WO2016085248A1 - Method for culturing natural killer cells using t cells - Google Patents

Method for culturing natural killer cells using t cells Download PDF

Info

Publication number
WO2016085248A1
WO2016085248A1 PCT/KR2015/012700 KR2015012700W WO2016085248A1 WO 2016085248 A1 WO2016085248 A1 WO 2016085248A1 KR 2015012700 W KR2015012700 W KR 2015012700W WO 2016085248 A1 WO2016085248 A1 WO 2016085248A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
natural killer
cell
seed
killer cells
Prior art date
Application number
PCT/KR2015/012700
Other languages
French (fr)
Inventor
Bo Kyung Min
Hana Choi
Yu Kyeong Hwang
Original Assignee
Green Cross Lab Cell Corporation
Mogam Biotechnology Institute
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Green Cross Lab Cell Corporation, Mogam Biotechnology Institute filed Critical Green Cross Lab Cell Corporation
Priority to US15/527,752 priority Critical patent/US20170319621A1/en
Priority to EP15862930.3A priority patent/EP3224349B1/en
Priority to EP20216695.5A priority patent/EP3878950A1/en
Priority to JP2017525394A priority patent/JP6720168B2/en
Priority to CN201580063858.5A priority patent/CN107002039B/en
Priority to ES15862930T priority patent/ES2860974T3/en
Priority to AU2015354941A priority patent/AU2015354941B2/en
Publication of WO2016085248A1 publication Critical patent/WO2016085248A1/en
Priority to US17/220,865 priority patent/US11766456B2/en
Priority to US18/455,003 priority patent/US20240050478A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4613Natural-killer cells [NK or NK-T]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0646Natural killers cells [NK], NKT cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K2035/124Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells the cells being hematopoietic, bone marrow derived or blood cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/47Brain; Nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/48Blood cells, e.g. leukemia or lymphoma
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/53Liver
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/59Reproductive system, e.g. uterus, ovaries, cervix or testes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2302Interleukin-2 (IL-2)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/50Cell markers; Cell surface determinants
    • C12N2501/515CD3, T-cell receptor complex
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2502/00Coculture with; Conditioned medium produced by
    • C12N2502/11Coculture with; Conditioned medium produced by blood or immune system cells
    • C12N2502/1114T cells

Definitions

  • the present invention relates to a method for producing natural killer cells using T cells, and more particularly, to a method for producing natural killer cells, which comprises culturing seed cells using CD4(+) T cells as feeder cells.
  • Natural killer cells are known as lymphoid cells that account for about 10-15% of peripheral blood lymphocytes and play an important role in innate immune responses. Unlike T cells, natural killer cells recognize their target in an MHC non-restricted manner, and activating receptors (such as NKG2D, NCR (NKp30, NKp44, or NKp46)) in natural killer cells compete with inhibitory receptors such as KIR or CD94/NKG2A to display activity and eliminate tumor target cells. Natural killer cells can exhibit antiviral effects, an-GvH effects, and anticancer effects.
  • natural killer cells directly kill malignant tumors, including sarcoma, myeloma, carcinoma, lymphomas and leukemia, or contribute to adaptive immune activation by inducing dendritic cell (DC) activity or tumor-specific cytotoxic T lymphocytes (CTLs), thereby eliminating abnormal cells which are tumor cells or cells developing into tumor cells.
  • DC dendritic cell
  • CTLs tumor-specific cytotoxic T lymphocytes
  • the anticancer effects of natural killer cells were demonstrated through allogeneic hematopoietic stem cell transplantation, and it was found that donor natural killer cells inhibit microtumor remaining after transplantation of T cell-depleted hematopoietic stem cells.
  • donor natural killer cells inhibit microtumor remaining after transplantation of T cell-depleted hematopoietic stem cells.
  • graft-versus-tumor (GVT) effect of donor natural killer cells significantly increases when there is KIR (killer cell immunoglobulin-like receptors)-MHC mismatch between the donor and the recipient, the allogeneic natural killer cells is much more effective than the use of the cancer patient’s own autologous natural killer cells having reduced function.
  • PBMC In ex vivo expansion culture of natural killer cells, PBMC, CD3- cells, CD3-CD56+ cells, CD56+ cells or the like are used as seed cells, and cytokines such as IL-2, IL-12, IL-15 or IL-21, and OKT-3 antibody (Condiotti et al., Experimental Hematol . 29(1):104-113, 2001) stimulating LPS (Goodier et al., J. Immunol . 165(1):139-147, 2000) and CD3, are used as growth factors for natural killer cells.
  • cytokines such as IL-2, IL-12, IL-15 or IL-21
  • OKT-3 antibody Condiotti et al., Experimental Hematol . 29(1):104-113, 2001
  • stimulating LPS Goodier et al., J. Immunol . 165(1):139-147, 2000
  • CD3 In ex vivo expansion culture of natural killer cells, PBMC, CD3- cells, CD3-CD
  • PBMCs stimulated with OKT-3 can be used as feeder cells, and also disclosed that, when the PBMCs were cultured for 14 days, a 691-fold increase in cell number could be induced (Lim et al., PlosOne , 7(1):e53611, 2012).
  • the present inventors have disclosed that, when PBMCs were re-stimulated twice or more with feeder cells, an increase of thousands to tens of thousands of times in cell number could be induced (WO2013/094988).
  • T lymphocytes particularly helper T cells (Th cells)
  • Th cells helper T cells
  • Another object of the present invention is to provide natural killer cells produced by the above method, a composition for preventing or treating cancer or infectious disease, which comprises the natural killer cells as an active ingredient, and a method for preventing or treating cancer or infectious disease, comprising administering to a subject in need thereof a therapeutically effective amount of the above composition.
  • the present invention provides a method for producing natural killer cells, wherein T cells, particularly CD4(+) T cells, are used as feeder cells that stimulate the natural killer cells.
  • the present invention also provides natural killer cells produced by the above method.
  • the present invention also provides a composition for preventing or treating cancer or infectious disease, which comprises the above natural killer cells as an active ingredient.
  • the present invention also provides a method for preventing or treating cancer or infectious disease, administering to a subject in need thereof a therapeutically effective amount of the above composition.
  • FIG. 1a shows the results of measuring the increase in the number of total nucleated cells and natural killer cells, obtained by culturing PBMC seed cells using various T cells as feeder cells for 14 days (condition 1 in Table 1 below).
  • FIG. 1b shows the results of measuring the increase in the number of total nucleated cells and natural killer cells, obtained by culturing PBMC seed cells using various T cells as feeder cells, and then re-stimulating the cells with various T cells serving as feeder cells, and culturing the cells up to day 21 (condition 2 in Table 1 below).
  • FIG. 1c shows the results of measuring the increase in the number of total nucleated cells and natural killer cells, obtained by culturing CD3(+) cell-depleted PBMC seed cells using various T cells as feeder cells for 14 days (condition 3 in Table 1 below).
  • FIG. 1d shows the results of measuring the increase in the number of total nucleated cells and natural killer cells, obtained by culturing CD3(+) cell-depleted PBMC seed cells using PBMC feeder cells, and then re-stimulating the cells with various T cells serving as feeder cells, and culturing the cells up to day 21 (condition 4 in Table 1 below).
  • FIG. 1e shows the results of measuring the increase in the number of total nucleated cells of poorly proliferating donors, cultured under condition 3 of Table 1 below.
  • FIG. 2 shows the cell viabilities of natural killer cells cultured using various T cells as feeder cells.
  • FIG. 3a shows the identity and purity of natural killer cells obtained by culturing PBMC seed cells using various T cells as feeder cells for 14 days (condition 1 in Table 1 below).
  • FIG. 3b shows the identity and purity of natural killer cells obtained by culturing PBMC seed cells using various T cells as feeder cells, and then re-stimulating the cells with various T cells serving as feeder cells, and culturing the cells up to day 21 (condition 2 in Table 1 below).
  • FIG. 3c shows the identity and purity of natural killer cells obtained by culturing CD3(+) cell-depleted PBMC seed cells using various T cells as feeder cells for 14 days (condition 3 in Table 1 below).
  • FIG. 3d shows the identity and purity of natural killer cells obtained by culturing CD3(+) cell-depleted PBMC seed cells using PBMC feeder cells, and then re-stimulating the cells with various T cells serving as feeder cells, and culturing the cells up to day 21 (condition 4 in Table 1 below).
  • FIG. 3e shows the identity and purity of natural killer cells of poorly proliferating donors, obtained by culture under condition 3 of Table 1 below.
  • FIG. 4a shows an activation marker in natural killer cells obtained by culturing PBMC seed cells using various T cells as seed cells for 14 days (condition 1 in Table 1 below 1).
  • FIG. 4b shows an activation marker in natural killer cells obtained by CD3(+) cell-depleted PBMC seed cells using various T cells as feeder cells for 14 days (condition 3 in Table 1 below).
  • FIG. 5a shows the cell killing activities against various cancers of natural killer cells obtained by culturing PBMC seed cells using various T cells as feeder cells for 14 days (condition 1 in Table 1 below).
  • FIG. 5b shows the cell killing activities against various cancers of natural killer cells obtained by culturing PBMC seed cells using various T cells as feeder cells, and then re-stimulating the cells with various T cells serving as feeder cells, and culturing the cells up to day 21 (condition 2 in Table 1 below).
  • FIG. 5c shows the cell killing activities against various cancers of natural killer cells obtained by culturing CD3(+) cell-depleted PBMC seed cells using various T cells as feeder cells for 14 days (condition 3 in Table 1 below).
  • FIG. 5d shows the cell killing activities against various cancers of natural killer cells obtained by culturing CD3(+) cell-depleted PBMC seed cells using PBMC feeder cells, and then re-stimulating the cells with various T cells serving as feeder cells, and culturing the cells up to day 21 (condition 4 in Table 1 below).
  • FIG. 5e shows the cell killing activities against K562 of natural killer cells of poorly proliferating donors, obtained by culture under condition 3 of Table 1 below.
  • T cells in a process of producing natural killer cells by proliferating the cells using various T cells as feeder cells, T cells, particularly CD4(+) T cells, induce the selective proliferation of natural killer cells, and thus the proliferation of the natural killer cells increases and the cell killing activity thereof also increases.
  • the present invention is directed to a method for producing natural killer cells, wherein CD4(+) T cells are used as feeder cells that stimulate the natural killer cells.
  • the present invention is directed to a method for producing natural killer cells, wherein CD4(+) T cells isolated ex vivo , CD4(+) T cells cultured ex vivo , or CD4(+) T cell lines are used as feeder cells, and one or more types of cells selected from the group consisting of peripheral blood cells, peripheral blood leukocyte cells, peripheral blood mononuclear cells (PBMCs), enriched natural killer cells and isolated natural killer cells are used as seed cells.
  • PBMCs peripheral blood mononuclear cells
  • the method for producing natural killer cells according to the present invention may concretely comprise the following steps, but is not limited thereto:
  • peripheral blood leukocyte cells peripheral blood mononuclear cells (PBMCs), T cell-depleted mononuclear cells or natural killer cells from human peripheral blood;
  • PBMCs peripheral blood mononuclear cells
  • Natural killer (NK) cells are present in an amount of about 10-15% in the blood of normal people, and have high killing ability when they react with non-self material. Natural killer cells non-specifically and immediately act in response to the infection of cells with various viruses, the penetration of bacteria or the production of abnormal cells to thereby remove foreign matter.
  • the number of natural killer cells present in the body is not so large, and the number of effective natural killer cells required to exhibit therapeutic effects should be very large. For this reason, a method for the proliferation and production of effective natural killer cells is required.
  • Methods for proliferating natural killer cells include a method comprising isolating natural killer cells only, suitably stimulating the isolated natural killer cells with feeder cells, and proliferating the stimulated natural killer cells, and a method comprising selectively proliferating natural killer cells from peripheral blood lymphocytes (PBLs) or peripheral blood mononuclear cells (PBMCs) to thereby obtain a relatively large amount of natural killer cells.
  • PBLs peripheral blood lymphocytes
  • PBMCs peripheral blood mononuclear cells
  • PBMCs are separated into lymphocytes and monocytes, and the lymphocytes are further divided into T cells, B cells, and natural killer cells. Among them, the T cells are further divided into helper T cells (Th cells) and cytotoxic T cells (Tc cells).
  • Th cells helper T cells
  • Tc cells cytotoxic T cells
  • feeder cells refers to cells that produce various metabolites due to their metabolic activity to thereby assist in the proliferation of target cells, even though these cells cannot proliferate.
  • Feeder cells that are used in the present invention may be CD4(+) T cells isolated ex vivo , CD4(+) T cells expansion-cultured ex vivo , or a CD4(+) T cell line (T lymphoma cell line).
  • the CD4(+) T cell line (T lymphoma cell line) that is used in the present invention is concretely H9, HuT78, Loucy, Molt3, Molt-13, PEER, RPMI8402, or TALL-01, more concretely H9 or HuT78, but is not limited thereto.
  • T cells that are used as the feeder cells may be inactivated or non-inactivated cells whose proliferation was inhibited. Concretely, the T cells may be inactivated to ensure their safety.
  • a method for inactivating the T cells a conventional method known in the art may be used, and for example, a method of irradiating the T cells with gamma-rays may be used. If non-inactivated T cells are used, they can be killed by natural killer cells during culture, because they are mostly tumor cells.
  • the proliferation method as described in the present invention uses T cells as feeder cells.
  • This method has an advantage in that, because the culture of natural killer cells is selectively induced in seed cells such as PBMCs without removing T cells, the culture can be stably performed without a significant difference in culture results between donors. Thus, it is possible to obtain an increased amount of natural killer cells for therapeutic purposes in an efficient and stable manner.
  • seed cells means cells capable of proliferating to natural killer cells by suitable culture.
  • the seed cells that are used in the present invention may be one or more types selected from the group consisting of peripheral blood cells, peripheral blood leukocytes, PBMCs (peripheral blood mononuclear cells), enriched natural killer cells, and isolated natural killer cells, but are not limited thereto. More concretely, the seed cells may be CD3(+) cell-depleted cells(CD3(-) cells).
  • natural killer cells are concretely cultured in a medium containing an interleukin protein and a T cell-stimulating antibody having a low affinity for T cells, but the scope of the present invention is not limited thereto.
  • the T cell-stimulating antibody having a low affinity for T cells is a protein that reacts specifically with CD3 antigen, a group of molecules which associate with a T cell receptor (TCR) to form an antigen recognition complex, in which the CD3 molecule has a longer intracellular domain compared to TCR and functions to transfer an antigen recognition signal into cells.
  • the T cell-stimulating antibody having a low affinity for T cells which can be used in the present invention, is concretely anti-CD3 antibody, more concretely OKT-3, UCHT1, HIT3a or the like, most concretely OKT-3 antibody.
  • interleukin (IL) protein refers to a collection of biologically active proteins produced by immune cells such as lymphocytes, monocytes or macrophages, and means a group of molecular species in cytokines.
  • examples of an interleukin protein which may be used in the present invention, include IL-2, IL-15, IL-12, IL-18, IL-21 and the like. Concretely, the interleukin protein is IL-2 protein.
  • the method for producing natural killer cells concretely comprises: adding natural killer cells and a T lymphoma cell line to a conventional animal cell culture medium such as AIM-V medium, RIMI 1640, CellGro SCGM, X-VIVO20, IMDM or DMEM; adding an interleukin protein and T cell-stimulating antibody having a low affinity for T cells to the culture medium; and culturing the cells, but is not limited thereto.
  • a conventional animal cell culture medium such as AIM-V medium, RIMI 1640, CellGro SCGM, X-VIVO20, IMDM or DMEM
  • adding an interleukin protein and T cell-stimulating antibody having a low affinity for T cells to the culture medium
  • cells were cultured in a medium containing OKT-3 antibody and IL-2.
  • OKT-3 antibody is used at a concentration of 0.1-100ng/ml, concretely about 10ng/ml, and IL-2 is used at a concentration of 10-2000U/ml, concretely about 500U/ml.
  • the culture medium may further contain serum or plasma and an additional growth factor that supports the proliferation of lymphocytes.
  • the type of serum or plasma that is added to the medium is not specifically limited, and thus various types of commercially available serum or plasma may be used in the present invention. Concretely, autologous human serum or plasma is used in the present invention.
  • the method for producing natural killer cells comprises a step of culturing natural killer cells in a medium containing an interleukin protein and a T cell-stimulating antibody having a low affinity for T cells, in the presence of T cells as feeder cells that stimulate natural killer cells.
  • the feeder cells and the seed cells are concretely mixed at a ratio of at least 1:1, more concretely 2:1-20:1, most concretely 5:1, but are not limited thereto.
  • ratio refers to a ratio based on cell number.
  • the stimulation and culture of natural killer cells with CD4(+) T cells serving as feeder cells may be repeatedly performed in order to obtain an increased number of natural killer cells.
  • the method of the present invention may further comprise re-stimulating natural killer cells at a feeder cell-to-seed cell ratio of at least 1:1, concretely 2:1-20:1, most concretely 5:1.
  • ratio refers to a ratio based on cell number.
  • the CD4(+) T cells that are used in the re-stimulation are concretely H9 or HuT78 cells, but are not limited thereto.
  • inactivated cells whose proliferation was inhibited, or non-inactivated cells may be used in the re-stimulation.
  • the re-stimulation is concretely performed in a medium containing an interleukin protein and a T cell-stimulating antibody having a low affinity for T cells, at intervals of 5-12 days, more concretely 7 days, but is not limited, and may be repeated at least once.
  • the method of the present invention may further comprise, after the step of culturing the cells in the medium containing the interleukin protein and the T cell-stimulating antibody having a low affinity for T cells, a step of culturing the cells in a medium free of the antibody.
  • the T cell-stimulating antibody having a low affinity for T cells may be an anti-CD3 antibody, concretely at least one selected from the group consisting of OKT3, UCHT1 and HIT3a antibodies, most concretely OKT3 antibody, and the interleukin protein may be at least one selected from the group consisting of IL-2, IL-12, IL-15, IL-18, and IL-21, concretely IL-2.
  • the term “stimulating” means inducing the proliferation of natural killer cells by adding feeder cells or the like.
  • a T cell-stimulating antibody having a low affinity for T cells may also be used.
  • the term “re-stimulating” means re-inducing the proliferation of natural killer cells by adding feeder cells and/or a T cell-stimulating antibody, which has a low affinity for T cells, to the medium after a certain time of culture.
  • the natural killer cells are cultured for at least 5 days, concretely 5-60 days, more concretely 14-21 days, but are not limited thereto.
  • the stimulation may start at day 0 of culture and may be repeated at intervals of 5-12 days, concretely 7 days, but is not limited thereto.
  • the cells may be harvested at 5 days or more, concretely 14 days, after the final stimulation, but are not limited thereto.
  • Natural killer cells cultured according to the method of the present invention as described above can be frozen and are not functionally impaired even when they are thawed.
  • these cells express higher levels of activating receptors such as NKp44 and NKp46 compared to conventional cells cultured using PBMCs as feeder cells, and thus have increased abilities to kill tumor cells and to secrete cytokines, indicating that these cells can exhibit excellent anticancer effects.
  • a cell therapeutic agent effective for tumor treatment can be prepared using a large amount of activated natural killer cells that can be clinically applied.
  • Example 1 Preparation of PBMCs and Various T Cells
  • PBMCs Peripheral blood mononuclear cells
  • PBS phosphate buffered saline
  • FBS fetal serum bovine
  • the PBMC pellets were suspended in 10mL of MACS running buffer, and the cells were counted using an Adam cell counter system.
  • PBMC seed cells 1 ⁇ 10 7 PBMC seed cells were dispensed into each of 50mL tubes.
  • PBMC feeder cells and CD3(+) cell-depleted seed cells 5 ⁇ 10 7 cells were transferred into each of 50mL fresh tubes and centrifuged at 1200 rpm at 4°C for 10 minutes.
  • the cell pellets were suspended in 10mL of 1 vol % autoplasma-containing CellGro medium (Cellgenix).
  • CD3(+)-depleted seed cells 400 ⁇ L of MACS running buffer and 100 ⁇ L of CD3 magnetic beads (Miltenyi biotech) were added to 5 ⁇ 10 7 PBMC cell pellets which were then incubated at 4°C for 20 minutes. The resulting cells were washed with 10-20mL of MACS running buffer, and then centrifuged at 1200 rpm at 4°C for 10 minutes and suspended aging in 2mL of MACS running buffer. The cells were separated using a CS column (Miltenyi Biotech, 130-041-305) equipped with VarioMACS (Miltenyi Biotech), and the column was washed to reach a final volume of 20mL, thereby recovering the cells.
  • the cells were counted using an Adam cell counter system, and 1 ⁇ 10 7 cells were dispensed into 50mL fresh tubes and centrifuged at 1200 rpm at 4°C for 10 minutes.
  • the cell pellets were suspended in 10mL of CellGro medium (Cellgenix) containing 1 vol % autoplasma.
  • PBMC feeder cell pellets separated in Example 1-1 were suspended in 10mL of 1 vol % autoplasma-containing CellGro medium (Cellgenix) and irradiated with 2000 cGy of gamma rays in a gamma-ray irradiator, thereby preparing PBMC feeder cells.
  • autoplasma-containing CellGro medium Cellgenix
  • T cells were recovered from culture flasks and centrifuged at 1200 rpm at 4°C for 10 minutes, and then 5 ⁇ 10 7 cells were dispensed into each tube and centrifuged at 1200 rpm at 4°C for 10 minutes.
  • the T cells were suspended in 10mL of 1 vol % autoplasma-containing CellGro medium (Cellgenix), and then irradiated with 15000-30000 cGy of gamma rays in a gamma ray irradiator, thereby preparing T cells for use as feeder cells.
  • Example 2 Culture of Natural Killer Cells Using Various T Cells as Feeder Cells
  • a culture vessel a flask or a cell culture bag.
  • PBMC seed cells and PBMC feeder cells or T feeder cells were added to the culture vessel at a ratio of 1:5 in an amount of 0.5-10mL for each cell type, and 0.5-10mL of 1 vol % autoplasma-containing CellGro medium (Cellgenix) was added to the cells which were then stationary-cultured in an incubator at 37°C for 3-5 days (condition 1).
  • CD3(-) seed cells and PBMC feeder cells or T seed cells were added to a culture vessel at a ratio of 1:5 in an amount of 0.5-10 mL for each cell type, and 0.5-10mL of 1 vol % autoplasma-containing CellGro medium (Cellgenix) was added to the cells which were then stationary-cultured in an incubator at 37°C for 3-5 days (condition 3).
  • the cells were counted at days 3-5 of culture, and diluted to a density of about 2-5 ⁇ 10 5 cells/mL with CellGro medium (Cellgenix) containing 500IU of IL-2 (Proleukin) and 1 vol % autoplasma.
  • the diluted cells were stationary-cultured again in a suitable culture vessel.
  • the cells were suspension-cultured up to day 14 while the cells were counted at intervals of 2-3 days and diluted to a density of 5-10 ⁇ 10 5 cells/mL CellGro medium (Cellgenix) containing 500IU of IL-2 and 1 vol% autoplasma.
  • natural killer cells were harvested.
  • PBMC feeder cells showed a 147-fold increase in the number of total nucleated cells (TNCs), and H9 feeder cells and HuT78 feeder cells showed a 298-fold increase and a 485-fold increase, respectively, which were significantly higher than the fold increase shown by PBMC feeder cells.
  • PBMC feeder cells showed a 247-fold increase in the number of natural killer cells
  • H9 feeder cells showed a 2752-fold increase which was about 10 times higher than that shown by PBMC feeder cells
  • HuT78 feeder cells showed a 5649-fold increase which was about 20 times higher than that shown by PBMC feeder cells.
  • other T cells showed proliferation rates lower than that shown by PBMCs (see FIG. 1a).
  • PBMCs showed a 184-fold increase in the number of total nucleated cells
  • H9 feeder cells showed a 62-fold increase
  • HuT78 feeder cells showed a 217-fold increase (see FIG. 1c).
  • Other T cells all showed proliferation rates lower than that shown by H9.
  • a culture vessel a flask or a cell culture bag.
  • PBMC seed cells and PBMC feeder cells or T feeder cells were added to the culture vessel at a ratio of 1:5 in an amount of 0.5-10mL for each cell type, and 0.5-10mL of 1 vol % autoplasma-containing CellGro medium (Cellgenix) was added to the cells which were then stationary-cultured in an incubator at 37°C for 3-5 days (condition 1).
  • the natural killer cells being cultured were counted and diluted to a density of 2-5 ⁇ 10 5 cells/mL with 1 vol% autoplasma-containing CellGro medium (Cellgenix), and PBMC and T feeder cells were diluted 3- to 10-fold and suspended in 1 vol % autoplasma-containing CellGro medium (Cellgenix), and then inactivated by irradiation with 2000 and 15000-30000 cGy of gamma-rays, respectively, in a gamma-ray irradiator. 500IU of IL-2 and 10ng/mL of OKT-3 were added to the medium, and the prepared two types of cells were co-cultured.
  • the cells were suspension-cultured up to day 21 while the cells were counted at intervals of 2-3 days and diluted to a density of 5-10 ⁇ 10 5 cells/mL with CellGro medium (Cellgenix) containing 500IU of IL-2 and 1 vol % autoplasma.
  • CellGro medium Cellgenix
  • natural killer cells were harvested.
  • the proliferation rates of natural killer cells cultured under condition 2 shown in Table 1 were compared. As a result, it was shown that the fold increase in the number of total nucleated cells was higher in the order of PBMCs (334-fold), H9 (358-fold) and HuT78 (2282-fold). The fold increase in the number of natural killer cells was 1257-fold for PBMCs, 2677-fold for H9, and 29455-fold for HuT78 (FIG. 1b).
  • H9 and HuT78 can be used as feeder cells that are very excellent in terms of proliferation rate compared to PBMCs.
  • the results of evaluation performed using CD3(-) cells as seed cells and various T cells as feeder cells did slightly differ from the results obtained using PBMCs as feeder cells. Specifically, the proliferation rate of natural killer cells was similar between the PBMC feeder cells and the HuT78 feeder cells, and H9 feeder cells showed a reduced proliferation rate of natural killer cells compared to the PBMC feeder cells.
  • Donor cells that showed poor proliferation when cultured with PBMC feeder cells under condition 4 of Table 1 were selected and used to compare the proliferation rates of natural killer cells.
  • T cells In the case of PBMC feeder cells, a process of depleting T cells from seed cells is necessarily required, because the proliferation of T cells rather than natural killer cells is induced when T cells are present in seed cells. In addition, a preculture process for previously selecting donors is required, because the difference in proliferation rate between donors is significant. However, it was found that T cells, particularly CD4-expressing T cells, could induce the selective proliferation of natural killer cells regardless of the presence or absence of T cells in seed cells and that the proliferation of poorly proliferating donor cells could also be easily induced when culture with PBMC feeder cells was performed.
  • an ADAM cell counter system was used to comparatively evaluate in vitro cell viabilities. The number of viable cells was determined by subtracting the measured dead cell count from the measured total cell count, and then cell viability was calculated using the following equation.
  • the viability of natural killer cells was measured after culture under conditions 1 and 2 (that is, 14-day culture and 21-day culture of PBMC seed cells with various feeder cells) shown in Table 1. As a result, it was shown that the use of PBMC feeder cells showed a viability lower than 80%, and H9 and HuT78 showed a viability of 80% or higher (FIGS. 2a and 2b).
  • the viability of natural killer cells was measured after culture under conditions 3 and 4 (that is, 14-day culture and 21-day culture of CD3(+) T cell-depleted seed cells with various T feeder cells) shown in Table 1 were measured. As a result, it was shown that a high viability of about 90% or higher was shown in all the conditions. In the case of poorly proliferating donor cells, PBMC feeder cells showed a viability of 82%, and HuT78 showed a viability of 93% which was at least 10% higher than that shown by PBMC feeder cells (FIGS. 2c and 2d).
  • the viability of the cells cultured using PBMCs and various T feeder cells for 14 days or 21 days was generally higher in CD3(-) seed cells than in PBMC seed cells, and showed no significant difference between the types of seed cells.
  • HuT78 showed a viability which was about 10% higher than that shown by PBMC feeder cells.
  • HuT78 cells are feeder cells for culturing natural killer cells, which are useful in terms of cell viability compared to PBMCs.
  • Natural killer cells cultured according to the methods of Examples 1 and 2 were collected before and after culture and centrifuged at 1200 rpm for 5 minutes, and the medium was removed by suction.
  • the cells were diluted with 1 mL of FACS buffer (2.5% FBS-containing PBS), counted, and then diluted to a density of 5 ⁇ 10 6 cells/mL with FACS buffer. 100L of the diluted cell solution was dispensed into each of 5mL FACS tubes (Falcon, 352052), and the phenotypes of the cells were analyzed using the following antibody:
  • Tube 1 anti-human CD3-FITC (BD Pharmingen, 555332), anti-human CD16-PE (BD Pharmingen, 555407), anti-human CD56-PE-Cy5 (BD Pharmingen, 555517)
  • Tube 2 anti-human CD14-FITC (BD Pharmingen, 555397), anti-human CD19-PE (BD Pharmingen, 555413), anti-human CD3-PE-Cy5 (BD Pharmingen, 555341)
  • Tube 3 anti-human CD3-FITC, anti-human NKG2A-PE (R&D system, FAB1059P), anti-human CD56-PE-Cy5
  • Tube 4 anti-human CD3-FITC, anti-human NKG2C-PE (R&D system, FAB138P), anti-humanCD56-PE-Cy5
  • Tube 5 anti-human CD3-FITC, anti-human NKG2D-PE (R&D system, FAB139P), anti-human CD56-PE-Cy5
  • Tube 6 anti-human CD3-FITC, anti-human NKp30-PE (BD Pharmingen, 558407), anti-human CD56-PE-Cy5
  • Tube 7 anti-human CD3-FITC, anti-human NKp44-PE (BD Pharmingen, 558563), anti-humanCD56-PE-Cy5
  • Tube 8 anti-human CD3-FITC, anti-human NKp46-PE (BD Pharmingen, 557991), anti-human CD56-PE-Cy5
  • Tube 9 anti-human CD3-FITC, anti-human DNAM-1-PE (BD Pharmingen, 559789), anti-humanCD56-PE-Cy5
  • Tube 10 anti-human CD3-FITC, anti-human CD25-PE (BD Pharmingen, 555432), anti-human CD56-PE-Cy5
  • Tube 11 anti-human CD3-FITC, anti-human CD62L-PE (eBioscience, 12-0629-42), anti-human CD56-PE-Cy5
  • Tube 12 anti-human CD3-FITC, anti-human CD69-PE (R&D systems, FAB23591P), anti-human CD56-PE-Cy5
  • Tube 13 anti-human CD3-FITC, anti-human CXCR3-PE (BD Pharmingen, 557185), anti-human CD56-PE-Cy5
  • Tube 14 anti-human CD3-FITC, anti-human CD57-PE (BD Pharmingen, 560844), anti-human CD56-PE-Cy5
  • Tube 15 anti-human CD3-FITC, PE mouse IgG1 k isotype control (BD Pharmingen, 555749), anti-human CD56-PE-Cy5
  • Tube 16 FITC mouse IgG1 k isotype control (BD Pharmingen, 555748), PE mouse IgG1 k isotype control (BD Pharmingen, 555749), PE-Cy5 mouse IgG1 k isotype control (BD Pharmingen)
  • the tubes were stained at a cold temperature for 30 minutes, and the stained cells were added to 2mL of FACS buffer and centrifuged at 1500 rpm for 5 minutes. The supernatant was removed, and the remaining cells were added again to 2 mL of FACS buffer and centrifuged at 1500 rpm for 5 minutes. Next, the supernatant was removed, and the remaining cells were suspended in 300 ⁇ L of FACS buffer, and the phenotypes thereof were analyzed using FACS LSRII Fortessa (Becton Dickinson), thereby determining the identity and purity of the cells.
  • FACS LSRII Fortessa Becton Dickinson
  • the content was expressed as the contents of CD3(-)CD56(+) cells and CD16(+)CD56(+) cells, and the purity was measured with CD3(+) for T cells, CD14(+) for monocytes, and CD19(+) for B cells.
  • 3 ⁇ 10 6 natural killer cells were dispensed into each of 15mL tubes and centrifuged, and the pellets were suspended in RPMI 1640-10% FBS medium at a desired ratio relative to the target tumor cell line.
  • 100 ⁇ L of each of the prepared target tumor cell line and natural killer cell line was dispensed into each well of a round-bottom 96-well plate, and each well was prepared in triplicate.
  • 100 ⁇ L of the stained tumor cell line and 100 ⁇ L of RPMI 1640-10% FBS medium were added.
  • 100 ⁇ L of the stained tumor cell line and 100 ⁇ L of 2% Triton-X 100 solution were added.
  • the cells were incubated in an incubator at 37°C for 4 hours under a light-shielding condition, and then the plate was centrifuged at 2000 rpm for 3 minutes. 100 ⁇ L of the supernatant was added to each well of a 96-well black plate, and the fluorescence value (OD 480/ 535 nm ) was measured using a fluorescence plate reader (Perkin Elmer, VICTOR X3). Based on the measured fluorescence value, the tumor cell killing activity of the natural killer cells was calculated using the following equation:
  • % of killing (Average fluorescence value of sample wells ⁇ average fluorescence value of spontaneous wells)/ ⁇ (average fluorescence value of maximum wells + A) ⁇ average fluorescence value of spontaneous wells ⁇ ⁇ 100
  • Natural killer cells cultured with various feeder cells were allowed to react with various tumor cell lines, and the direct tumor cell killing activity of the natural killer cells was measured.
  • the cell killing activity of natural killer cells cultured under condition 1 14-day culture of PBMC seed cells with various feeder cells shown in Table 1 was evaluated against the blood cancer cell line K562, the liver cancer cell line HuH-7, the lymphoma cell line HuT78, the brain tumor cell line U87-MG, the retinoblastoma cell line SNUOT-Rbl, the neuroblastoma cell line SK-N-SH and the ovarian cancer cell line OVCAR-3.
  • culture with PBMC feeder cells showed a higher cell killing activity against all the tumor targets compared with culture with H9 and HuT78 (FIG. 5a).
  • the cell killing activity of natural killer cells cultured under condition 3 was evaluated against the blood cancer cell line K562, the liver cancer cell line HuH-7, the lymphoma cell line HuT78, the brain tumor cell line U87-MG, the retinoblastoma cell line SNUOT-Rbl, the neuroblastoma cell line SK-N-SH and the ovarian cancer cell line OVCAR-3.
  • the cell killing activity of natural killer cells cultured under condition 4 (21-day culture of CD3(-) seed cells) shown in Table 1 was measured. As a result, it was shown that the cell killing activity against the K562 tumor cell line was similar between the types of feeder cells (FIG. 5d). This insignificant difference in specificity between the types of feeder cells appears to be because PBMCs were used in the first stimulation of seed cells in the same manner and PBMCs and various T cells were used only in the second stimulation.
  • HuT78 showed the highest cell killing activity against various tumor cell lines under most of the conditions. Particularly, natural killer cells cultured with H9 and HuT78 showed higher killing activities against tumor cell lines having resistance, indicating that H9 and HuT78 cells are feeder cells which are better than PBMCs in terms of efficacy.
  • the method for producing natural killer cells using T cells is a method capable of producing natural killer cells by selectively proliferating only natural killer cells from a small amount of seed cells while maintaining the high killing activity of the natural killer cells.
  • the method of the present invention can produce a large amount of natural killer cells that can be frozen, and thus is useful for commercialization of cell therapeutic agents.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Cell Biology (AREA)
  • Microbiology (AREA)
  • Hematology (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Oncology (AREA)
  • Virology (AREA)
  • Developmental Biology & Embryology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Communicable Diseases (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The present invention relates to a method for producing natural killer cells using T cells, and more particularly, to a method for producing natural killer cells, which comprises culturing seed cells using CD4(+) T cells as feeder cells. The method for producing natural killer cells using T cells according to the present invention is a method capable of producing natural killer cells by selectively proliferating only natural killer cells from a small amount of seed cells while maintaining the high killing activity of the natural killer cells. The method of the present invention can produce a large amount of natural killer cells that can be frozen, and thus is useful for commercialization of cell therapeutic agents.

Description

METHOD FOR CULTURING NATURAL KILLER CELLS USING T CELLS
The present invention relates to a method for producing natural killer cells using T cells, and more particularly, to a method for producing natural killer cells, which comprises culturing seed cells using CD4(+) T cells as feeder cells.
As therapy for preventing the metastasis and recurrence of cancer and extending the survival time of terminal cancer patients, immunotherapy based on the immune function of patients is receiving attention. In recent years, immunotherapy with genetically engineered T cells expressing antigen-specific chimeric antigen receptors (CARs) has been recognized as a promising approach for cancer therapy. However, when T cells of other people are used, they can cause series graft-versus-host disease (GVHD) due to major histocompatibility complex (MHC) restriction. For this reason, for commercialization of cell therapy, natural killer (NK) cells are much more useful, which can be used for allogeneic transplantation and are possible to produce in large amount and to freeze.
Natural killer cells are known as lymphoid cells that account for about 10-15% of peripheral blood lymphocytes and play an important role in innate immune responses. Unlike T cells, natural killer cells recognize their target in an MHC non-restricted manner, and activating receptors (such as NKG2D, NCR (NKp30, NKp44, or NKp46)) in natural killer cells compete with inhibitory receptors such as KIR or CD94/NKG2A to display activity and eliminate tumor target cells. Natural killer cells can exhibit antiviral effects, an-GvH effects, and anticancer effects. Particularly, natural killer cells directly kill malignant tumors, including sarcoma, myeloma, carcinoma, lymphomas and leukemia, or contribute to adaptive immune activation by inducing dendritic cell (DC) activity or tumor-specific cytotoxic T lymphocytes (CTLs), thereby eliminating abnormal cells which are tumor cells or cells developing into tumor cells.
The anticancer effects of natural killer cells were demonstrated through allogeneic hematopoietic stem cell transplantation, and it was found that donor natural killer cells inhibit microtumor remaining after transplantation of T cell-depleted hematopoietic stem cells. In addition, because the graft-versus-tumor (GVT) effect of donor natural killer cells significantly increases when there is KIR (killer cell immunoglobulin-like receptors)-MHC mismatch between the donor and the recipient, the allogeneic natural killer cells is much more effective than the use of the cancer patient’s own autologous natural killer cells having reduced function. Despite the potential of such natural killer cells as therapeutic agents against cancers or infectious diseases, most natural killer cells in the body of normal people exist in a resting state, and natural killer cells in the body of cancer patients lack their function due to the immune escape mechanism of cancer cells. In order to actually use natural killer cells as therapeutic agents, activated natural killer cells capable of recognizing and destroying tumor cells are required, and for this reason, it is very important to activate natural killer cells by ex vivo expansion culture from normal culture or patient’s blood. In addition, because the number of natural killer cells in vivo is limited, it is necessarily required to develop a technology capable of producing and freezing natural killer cells in large amounts that can sufficiently exhibit anticancer effects.
In ex vivo expansion culture of natural killer cells, PBMC, CD3- cells, CD3-CD56+ cells, CD56+ cells or the like are used as seed cells, and cytokines such as IL-2, IL-12, IL-15 or IL-21, and OKT-3 antibody (Condiotti et al., Experimental Hematol. 29(1):104-113, 2001) stimulating LPS (Goodier et al., J. Immunol . 165(1):139-147, 2000) and CD3, are used as growth factors for natural killer cells. However, the use of the above-mentioned growth factors alone allows natural killer cells to increase about 3-10-fold and cannot achieve sufficient proliferation. For this reason, in several studies, there was an attempt to proliferate natural killer cells using various types of feeder cells. It has been reported that the use of the leukemia cell line CTV-1 showed little or no improvement in proliferation (North et al., J. Immunol. 178(1):85-94, 2007), and culture with EBV-LCL for 21 days resulted in an increase in cell number of an average of about 490-fold (Berg et al., Cytotherapy, 11(3):341-355, 2009). It has been reported that, when culture was performed for 7 days to 3 weeks using artificial APCs (antigen presenting cells) obtained by expressing 4-1 BBL and membrane-bound IL-15 in the K562 cell line, cell number increased an average of 90-209-fold (Fujisaki et al., Cancer Res. 69(9):4010-4017, 2009). In addition, it has been reported that when a K562 cell line having the MICA, 4-1BBL and IL-15 expressed therein was cultured for 3 weeks, cell number increased an average of 350-fold (Gong et al., Tissue Antigens, 76(6):467-475, 2010), and when a K562 cell line having membrane-bound IL-15 expressed therein was cultured for 3 weeks while the cells were re-stimulated at 7-day intervals, cell number increased an average of 21,000-fold (Denman et al., PlosOne , 7(1):e30264, 2012). In addition, it has been reported that, when PBMCs were cultured for 14 days using KL-1 (human T lymphoblast) and EBV-transformed B cells as feeder cells, an average of a 740-fold increase in the number of natural killer cells was induced (Lim et al., Cancer Res., 73(8):2598-6607, 2013).
In Korean Patent No. 10-1133185, the present inventors have disclosed that PBMCs stimulated with OKT-3 can be used as feeder cells, and also disclosed that, when the PBMCs were cultured for 14 days, a 691-fold increase in cell number could be induced (Lim et al., PlosOne, 7(1):e53611, 2012). In addition, the present inventors have disclosed that, when PBMCs were re-stimulated twice or more with feeder cells, an increase of thousands to tens of thousands of times in cell number could be induced (WO2013/094988). However, in the case of mass culture, the required amounts of PBMCs for use as feeder cells increases and the results of culture of natural killer cells change depending on the characteristics of each donor, and for these reasons, it is actually difficult to achieve the smooth supply and mass culture of raw materials for commercialization and the management of donors.
Accordingly, the present inventors have made extensive efforts to develop seed cells capable of substituting for PBMCs, and as a result, have found that among PBMCs, T lymphocytes, particularly helper T cells (Th cells), are very important in the proliferation of natural killer cells, and T-cell leukemia-lymphoma cell lines which can proliferate while having the characteristics of Th cells can selectively induce the culture of natural killer cells and allow natural killer cells to stably proliferate, thereby completing the present invention.
It is an object of the present invention to provide a method of producing natural killer cells by culturing and proliferating the cells in an efficient and stable manner.
Another object of the present invention is to provide natural killer cells produced by the above method, a composition for preventing or treating cancer or infectious disease, which comprises the natural killer cells as an active ingredient, and a method for preventing or treating cancer or infectious disease, comprising administering to a subject in need thereof a therapeutically effective amount of the above composition.
To achieve the above objects, the present invention provides a method for producing natural killer cells, wherein T cells, particularly CD4(+) T cells, are used as feeder cells that stimulate the natural killer cells.
The present invention also provides natural killer cells produced by the above method.
The present invention also provides a composition for preventing or treating cancer or infectious disease, which comprises the above natural killer cells as an active ingredient.
The present invention also provides a method for preventing or treating cancer or infectious disease, administering to a subject in need thereof a therapeutically effective amount of the above composition.
Brief Description of Drawings
FIG. 1a shows the results of measuring the increase in the number of total nucleated cells and natural killer cells, obtained by culturing PBMC seed cells using various T cells as feeder cells for 14 days (condition 1 in Table 1 below).
FIG. 1b shows the results of measuring the increase in the number of total nucleated cells and natural killer cells, obtained by culturing PBMC seed cells using various T cells as feeder cells, and then re-stimulating the cells with various T cells serving as feeder cells, and culturing the cells up to day 21 (condition 2 in Table 1 below).
FIG. 1c shows the results of measuring the increase in the number of total nucleated cells and natural killer cells, obtained by culturing CD3(+) cell-depleted PBMC seed cells using various T cells as feeder cells for 14 days (condition 3 in Table 1 below).
FIG. 1d shows the results of measuring the increase in the number of total nucleated cells and natural killer cells, obtained by culturing CD3(+) cell-depleted PBMC seed cells using PBMC feeder cells, and then re-stimulating the cells with various T cells serving as feeder cells, and culturing the cells up to day 21 (condition 4 in Table 1 below).
FIG. 1e shows the results of measuring the increase in the number of total nucleated cells of poorly proliferating donors, cultured under condition 3 of Table 1 below.
FIG. 2 shows the cell viabilities of natural killer cells cultured using various T cells as feeder cells. (a): cell viability of natural killer cells cultured under condition 1 shown in Table 1; (b): cell viability of natural killer cells cultured under condition 2 shown in Table 1; (c): cell viability of natural killer cells cultured under condition 3 shown in Table 1; (d): cell viability of natural killer cells cultured under condition 4 shown in Table 1; and (e) cell viability of natural killer cells of poorly proliferating donors, cultured under condition 2 shown in Table 1.
FIG. 3a shows the identity and purity of natural killer cells obtained by culturing PBMC seed cells using various T cells as feeder cells for 14 days (condition 1 in Table 1 below).
FIG. 3b shows the identity and purity of natural killer cells obtained by culturing PBMC seed cells using various T cells as feeder cells, and then re-stimulating the cells with various T cells serving as feeder cells, and culturing the cells up to day 21 (condition 2 in Table 1 below).
FIG. 3c shows the identity and purity of natural killer cells obtained by culturing CD3(+) cell-depleted PBMC seed cells using various T cells as feeder cells for 14 days (condition 3 in Table 1 below).
FIG. 3d shows the identity and purity of natural killer cells obtained by culturing CD3(+) cell-depleted PBMC seed cells using PBMC feeder cells, and then re-stimulating the cells with various T cells serving as feeder cells, and culturing the cells up to day 21 (condition 4 in Table 1 below).
FIG. 3e shows the identity and purity of natural killer cells of poorly proliferating donors, obtained by culture under condition 3 of Table 1 below.
FIG. 4a shows an activation marker in natural killer cells obtained by culturing PBMC seed cells using various T cells as seed cells for 14 days (condition 1 in Table 1 below 1).
FIG. 4b shows an activation marker in natural killer cells obtained by CD3(+) cell-depleted PBMC seed cells using various T cells as feeder cells for 14 days (condition 3 in Table 1 below).
FIG. 5a shows the cell killing activities against various cancers of natural killer cells obtained by culturing PBMC seed cells using various T cells as feeder cells for 14 days (condition 1 in Table 1 below).
FIG. 5b shows the cell killing activities against various cancers of natural killer cells obtained by culturing PBMC seed cells using various T cells as feeder cells, and then re-stimulating the cells with various T cells serving as feeder cells, and culturing the cells up to day 21 (condition 2 in Table 1 below).
FIG. 5c shows the cell killing activities against various cancers of natural killer cells obtained by culturing CD3(+) cell-depleted PBMC seed cells using various T cells as feeder cells for 14 days (condition 3 in Table 1 below).
FIG. 5d shows the cell killing activities against various cancers of natural killer cells obtained by culturing CD3(+) cell-depleted PBMC seed cells using PBMC feeder cells, and then re-stimulating the cells with various T cells serving as feeder cells, and culturing the cells up to day 21 (condition 4 in Table 1 below).
FIG. 5e shows the cell killing activities against K562 of natural killer cells of poorly proliferating donors, obtained by culture under condition 3 of Table 1 below.
Best Mode For Carrying Out The Invention
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the invention pertains. Generally, the nomenclature used herein and the experiment methods, which will be described below, are those well known and commonly employed in the art.
In the present invention, it was found that, in a process of producing natural killer cells by proliferating the cells using various T cells as feeder cells, T cells, particularly CD4(+) T cells, induce the selective proliferation of natural killer cells, and thus the proliferation of the natural killer cells increases and the cell killing activity thereof also increases.
In one aspect, the present invention is directed to a method for producing natural killer cells, wherein CD4(+) T cells are used as feeder cells that stimulate the natural killer cells.
Concretely, the present invention is directed to a method for producing natural killer cells, wherein CD4(+) T cells isolated ex vivo, CD4(+) T cells cultured ex vivo, or CD4(+) T cell lines are used as feeder cells, and one or more types of cells selected from the group consisting of peripheral blood cells, peripheral blood leukocyte cells, peripheral blood mononuclear cells (PBMCs), enriched natural killer cells and isolated natural killer cells are used as seed cells.
The method for producing natural killer cells according to the present invention may concretely comprise the following steps, but is not limited thereto:
(i) a step of isolating peripheral blood leukocyte cells, peripheral blood mononuclear cells (PBMCs), T cell-depleted mononuclear cells or natural killer cells from human peripheral blood;
(ii) a step of culturing natural killer cells in a medium containing an interleukin protein and a T cell-stimulating antibody having a low affinity for T cells, in the presence of inactivated or non-inactivated T cells;
(iii) a step of re-stimulating the natural killer cells with T cells in a medium containing an interleukin protein and a T cell-stimulating antibody having a low affinity for T cells, and further culturing the natural killer cells.
Hereinafter, the present invention will be described in further detail.
Natural killer (NK) cells are present in an amount of about 10-15% in the blood of normal people, and have high killing ability when they react with non-self material. Natural killer cells non-specifically and immediately act in response to the infection of cells with various viruses, the penetration of bacteria or the production of abnormal cells to thereby remove foreign matter. However, the number of natural killer cells present in the body is not so large, and the number of effective natural killer cells required to exhibit therapeutic effects should be very large. For this reason, a method for the proliferation and production of effective natural killer cells is required.
Methods for proliferating natural killer cells include a method comprising isolating natural killer cells only, suitably stimulating the isolated natural killer cells with feeder cells, and proliferating the stimulated natural killer cells, and a method comprising selectively proliferating natural killer cells from peripheral blood lymphocytes (PBLs) or peripheral blood mononuclear cells (PBMCs) to thereby obtain a relatively large amount of natural killer cells. To isolate natural killer cells from peripheral blood, a conventional method known to one skilled in the art may be used, and commercially available natural killer cells may also be purchased and used.
PBMCs are separated into lymphocytes and monocytes, and the lymphocytes are further divided into T cells, B cells, and natural killer cells. Among them, the T cells are further divided into helper T cells (Th cells) and cytotoxic T cells (Tc cells). In the method of culturing natural killer cells using PBMC feeder cells, the mass proliferation of T cells is induced, and the proliferation of natural killer cells is limited, and for this reason, an expensive complex step of removing T cells is required before or after culture. Accordingly, in the present invention, whether any cell group among the PBMC groups contributes to the proliferation of natural killer cells was examined. As a result, it was found that T cells contribute to the proliferation of natural killer cells, and among them, Th cells are very important in the proliferation of natural killer cells. Based on this finding, T cells that can proliferate while having the characteristics of Th cells were used as feeder cells capable of PBMCs in the culture of natural killer cells.
As used herein, the term "feeder cells" refers to cells that produce various metabolites due to their metabolic activity to thereby assist in the proliferation of target cells, even though these cells cannot proliferate.
Feeder cells that are used in the present invention may be CD4(+) T cells isolated ex vivo, CD4(+) T cells expansion-cultured ex vivo, or a CD4(+) T cell line (T lymphoma cell line). The CD4(+) T cell line (T lymphoma cell line) that is used in the present invention is concretely H9, HuT78, Loucy, Molt3, Molt-13, PEER, RPMI8402, or TALL-01, more concretely H9 or HuT78, but is not limited thereto.
T cells that are used as the feeder cells may be inactivated or non-inactivated cells whose proliferation was inhibited. Concretely, the T cells may be inactivated to ensure their safety. As a method for inactivating the T cells, a conventional method known in the art may be used, and for example, a method of irradiating the T cells with gamma-rays may be used. If non-inactivated T cells are used, they can be killed by natural killer cells during culture, because they are mostly tumor cells.
The proliferation method as described in the present invention uses T cells as feeder cells. This method has an advantage in that, because the culture of natural killer cells is selectively induced in seed cells such as PBMCs without removing T cells, the culture can be stably performed without a significant difference in culture results between donors. Thus, it is possible to obtain an increased amount of natural killer cells for therapeutic purposes in an efficient and stable manner.
As used herein, the term "seed cells" means cells capable of proliferating to natural killer cells by suitable culture. Concretely, the seed cells that are used in the present invention may be one or more types selected from the group consisting of peripheral blood cells, peripheral blood leukocytes, PBMCs (peripheral blood mononuclear cells), enriched natural killer cells, and isolated natural killer cells, but are not limited thereto. More concretely, the seed cells may be CD3(+) cell-depleted cells(CD3(-) cells).
In addition, in the method for producing natural killer cells according to the present invention, natural killer cells are concretely cultured in a medium containing an interleukin protein and a T cell-stimulating antibody having a low affinity for T cells, but the scope of the present invention is not limited thereto. Herein, the T cell-stimulating antibody having a low affinity for T cells is a protein that reacts specifically with CD3 antigen, a group of molecules which associate with a T cell receptor (TCR) to form an antigen recognition complex, in which the CD3 molecule has a longer intracellular domain compared to TCR and functions to transfer an antigen recognition signal into cells. The T cell-stimulating antibody having a low affinity for T cells, which can be used in the present invention, is concretely anti-CD3 antibody, more concretely OKT-3, UCHT1, HIT3a or the like, most concretely OKT-3 antibody.
As used herein, the term “interleukin (IL) protein” refers to a collection of biologically active proteins produced by immune cells such as lymphocytes, monocytes or macrophages, and means a group of molecular species in cytokines. Examples of an interleukin protein, which may be used in the present invention, include IL-2, IL-15, IL-12, IL-18, IL-21 and the like. Concretely, the interleukin protein is IL-2 protein.
The method for producing natural killer cells according to the present invention concretely comprises: adding natural killer cells and a T lymphoma cell line to a conventional animal cell culture medium such as AIM-V medium, RIMI 1640, CellGro SCGM, X-VIVO20, IMDM or DMEM; adding an interleukin protein and T cell-stimulating antibody having a low affinity for T cells to the culture medium; and culturing the cells, but is not limited thereto. In an example of the present invention, cells were cultured in a medium containing OKT-3 antibody and IL-2. In the present invention, OKT-3 antibody is used at a concentration of 0.1-100ng/ml, concretely about 10ng/ml, and IL-2 is used at a concentration of 10-2000U/ml, concretely about 500U/ml. In addition, the culture medium may further contain serum or plasma and an additional growth factor that supports the proliferation of lymphocytes. The type of serum or plasma that is added to the medium is not specifically limited, and thus various types of commercially available serum or plasma may be used in the present invention. Concretely, autologous human serum or plasma is used in the present invention.
The method for producing natural killer cells according to the present invention comprises a step of culturing natural killer cells in a medium containing an interleukin protein and a T cell-stimulating antibody having a low affinity for T cells, in the presence of T cells as feeder cells that stimulate natural killer cells. For use in the culturing step, the feeder cells and the seed cells are concretely mixed at a ratio of at least 1:1, more concretely 2:1-20:1, most concretely 5:1, but are not limited thereto. Herein, the term "ratio" refers to a ratio based on cell number.
In addition, in the method for producing natural killer cells according to the present invention, the stimulation and culture of natural killer cells with CD4(+) T cells serving as feeder cells may be repeatedly performed in order to obtain an increased number of natural killer cells. Thus, the method of the present invention may further comprise re-stimulating natural killer cells at a feeder cell-to-seed cell ratio of at least 1:1, concretely 2:1-20:1, most concretely 5:1. Herein, the term "ratio" refers to a ratio based on cell number.
The CD4(+) T cells that are used in the re-stimulation are concretely H9 or HuT78 cells, but are not limited thereto. In addition, inactivated cells whose proliferation was inhibited, or non-inactivated cells, may be used in the re-stimulation.
In addition, the re-stimulation is concretely performed in a medium containing an interleukin protein and a T cell-stimulating antibody having a low affinity for T cells, at intervals of 5-12 days, more concretely 7 days, but is not limited, and may be repeated at least once. The method of the present invention may further comprise, after the step of culturing the cells in the medium containing the interleukin protein and the T cell-stimulating antibody having a low affinity for T cells, a step of culturing the cells in a medium free of the antibody.
Herein, the T cell-stimulating antibody having a low affinity for T cells may be an anti-CD3 antibody, concretely at least one selected from the group consisting of OKT3, UCHT1 and HIT3a antibodies, most concretely OKT3 antibody, and the interleukin protein may be at least one selected from the group consisting of IL-2, IL-12, IL-15, IL-18, and IL-21, concretely IL-2.
As used herein, the term "stimulating" means inducing the proliferation of natural killer cells by adding feeder cells or the like. In the stimulation step, a T cell-stimulating antibody having a low affinity for T cells may also be used. As used herein, the term "re-stimulating" means re-inducing the proliferation of natural killer cells by adding feeder cells and/or a T cell-stimulating antibody, which has a low affinity for T cells, to the medium after a certain time of culture.
In the method of the present invention, the natural killer cells are cultured for at least 5 days, concretely 5-60 days, more concretely 14-21 days, but are not limited thereto.
The stimulation may start at day 0 of culture and may be repeated at intervals of 5-12 days, concretely 7 days, but is not limited thereto. The cells may be harvested at 5 days or more, concretely 14 days, after the final stimulation, but are not limited thereto.
Natural killer cells cultured according to the method of the present invention as described above can be frozen and are not functionally impaired even when they are thawed. In addition, these cells express higher levels of activating receptors such as NKp44 and NKp46 compared to conventional cells cultured using PBMCs as feeder cells, and thus have increased abilities to kill tumor cells and to secrete cytokines, indicating that these cells can exhibit excellent anticancer effects. Thus, according to the present invention, a cell therapeutic agent effective for tumor treatment can be prepared using a large amount of activated natural killer cells that can be clinically applied.
EXAMPLES
Hereinafter, the present invention will be described in further detail with reference to examples. It will be obvious to a person having ordinary skill in the art that these examples are illustrative purposes only and are not to be construed to limit the scope of the present invention. Thus, the substantial scope of the present invention will be defined by the appended claims and equivalents thereof.
Example 1: Preparation of PBMCs and Various T Cells
1-1: Preparation of PBMC Seed Cells and CD3(+)-Depleted Seed Cells
Peripheral blood mononuclear cells (PBMCs) were dispensed into vials and frozen in liquid nitrogen. One frozen PBMC vial was thawed and transferred into a 50mL tube, and the cells were suspended in 20mL of PBS (phosphate buffered saline) containing 1 vol % FBS (fetal serum bovine) or autoplasma and were centrifuged at 1200 rpm at 4℃ for 10 minutes. The PBMC pellets were suspended in 10mL of MACS running buffer, and the cells were counted using an Adam cell counter system.
1×107 PBMC seed cells were dispensed into each of 50mL tubes. To obtain PBMC feeder cells and CD3(+) cell-depleted seed cells, 5×107 cells were transferred into each of 50mL fresh tubes and centrifuged at 1200 rpm at 4℃ for 10 minutes. In the case of PBMC feeder cells, the cell pellets were suspended in 10mL of 1 vol % autoplasma-containing CellGro medium (Cellgenix).
To obtain CD3(+)-depleted seed cells, 400μL of MACS running buffer and 100μL of CD3 magnetic beads (Miltenyi biotech) were added to 5×107 PBMC cell pellets which were then incubated at 4℃ for 20 minutes. The resulting cells were washed with 10-20mL of MACS running buffer, and then centrifuged at 1200 rpm at 4℃ for 10 minutes and suspended aging in 2mL of MACS running buffer. The cells were separated using a CS column (Miltenyi Biotech, 130-041-305) equipped with VarioMACS (Miltenyi Biotech), and the column was washed to reach a final volume of 20mL, thereby recovering the cells. The cells were counted using an Adam cell counter system, and 1×107 cells were dispensed into 50mL fresh tubes and centrifuged at 1200 rpm at 4℃ for 10 minutes. The cell pellets were suspended in 10mL of CellGro medium (Cellgenix) containing 1 vol % autoplasma.
1-2: PBMC Feeder Cells and Various T Feeder Cells
5×107 PBMC feeder cell pellets separated in Example 1-1 were suspended in 10mL of 1 vol % autoplasma-containing CellGro medium (Cellgenix) and irradiated with 2000 cGy of gamma rays in a gamma-ray irradiator, thereby preparing PBMC feeder cells.
Various T cells were recovered from culture flasks and centrifuged at 1200 rpm at 4℃ for 10 minutes, and then 5×107 cells were dispensed into each tube and centrifuged at 1200 rpm at 4℃ for 10 minutes. The T cells were suspended in 10mL of 1 vol % autoplasma-containing CellGro medium (Cellgenix), and then irradiated with 15000-30000 cGy of gamma rays in a gamma ray irradiator, thereby preparing T cells for use as feeder cells.
Example 2: Culture of Natural Killer Cells Using Various T Cells as Feeder Cells
Conditions for culturing PBMC seed cells and CD3(+)-depleted seed cells using T cells as feeder cells are shown in Table 1 below.
Figure PCTKR2015012700-appb-T000001
2-1: 14-Day Culture Conditions
For culture of natural killer cells, 500 IU of IL-2 and 10ng/mL of OKT-3 were added to a culture vessel (a flask or a cell culture bag). At day 0 of culture, PBMC seed cells and PBMC feeder cells or T feeder cells were added to the culture vessel at a ratio of 1:5 in an amount of 0.5-10mL for each cell type, and 0.5-10mL of 1 vol % autoplasma-containing CellGro medium (Cellgenix) was added to the cells which were then stationary-cultured in an incubator at 37℃ for 3-5 days (condition 1). In addition, at day 0 of culture, CD3(-) seed cells and PBMC feeder cells or T seed cells were added to a culture vessel at a ratio of 1:5 in an amount of 0.5-10 mL for each cell type, and 0.5-10mL of 1 vol % autoplasma-containing CellGro medium (Cellgenix) was added to the cells which were then stationary-cultured in an incubator at 37℃ for 3-5 days (condition 3).
In the case of conditions 1 and 3 shown in Table 1, the cells were counted at days 3-5 of culture, and diluted to a density of about 2-5×105 cells/mL with CellGro medium (Cellgenix) containing 500IU of IL-2 (Proleukin) and 1 vol % autoplasma. The diluted cells were stationary-cultured again in a suitable culture vessel. Next, the cells were suspension-cultured up to day 14 while the cells were counted at intervals of 2-3 days and diluted to a density of 5-10×105 cells/mL CellGro medium (Cellgenix) containing 500IU of IL-2 and 1 vol% autoplasma. At day 14 of suspension culture, natural killer cells were harvested.
The proliferation rates of the natural killer cells cultured under condition 1 were compared, and as a result, it was shown that PBMC feeder cells showed a 147-fold increase in the number of total nucleated cells (TNCs), and H9 feeder cells and HuT78 feeder cells showed a 298-fold increase and a 485-fold increase, respectively, which were significantly higher than the fold increase shown by PBMC feeder cells. Also, in the proliferation rate of natural killer cells among total nucleated cells, PBMC feeder cells showed a 247-fold increase in the number of natural killer cells, H9 feeder cells showed a 2752-fold increase which was about 10 times higher than that shown by PBMC feeder cells, and HuT78 feeder cells showed a 5649-fold increase which was about 20 times higher than that shown by PBMC feeder cells. In addition, other T cells showed proliferation rates lower than that shown by PBMCs (see FIG. 1a).
In addition, the proliferation rates of natural killer cells cultured under condition 3 were compared, and as a result, PBMCs showed a 184-fold increase in the number of total nucleated cells, H9 feeder cells showed a 62-fold increase, and HuT78 feeder cells showed a 217-fold increase (see FIG. 1c). Other T cells all showed proliferation rates lower than that shown by H9.
2-2: Re-stimulation Conditions in 21-Day Culture
For culture of natural killer cells, 500IU of IL-2 and 10ng/mL of OKT-3 were added to a culture vessel (a flask or a cell culture bag). At day 0 of culture, PBMC seed cells and PBMC feeder cells or T feeder cells were added to the culture vessel at a ratio of 1:5 in an amount of 0.5-10mL for each cell type, and 0.5-10mL of 1 vol % autoplasma-containing CellGro medium (Cellgenix) was added to the cells which were then stationary-cultured in an incubator at 37℃ for 3-5 days (condition 1). For re-stimulation at day 7 of culture, natural killer cells, stimulated and cultured with PBMC feeder cells at day 0, were re-stimulated with PBMC feeder cells and various T feeder cells, and cells, stimulated and cultured with T feeder cells at day 0, were re-stimulated using the same cells as feeder cells (condition 2). In addition, at day 0 of culture, CD3(-) seed cells and PBMC feeder cells were added to a culture vessel at a ratio of 1:5 in an amount of 0.5-10mL for each cell type, and 0.5-10mL of 1 vol% autoplasma-containing CellGro medium was added to the cells which were then stationary-cultured in an incubator at 37℃ for 3-5 days, thereby achieving the first stimulation of natural killer cells. In this case, for re-stimulation at day 7 of culture, natural killer cells were re-stimulated with PBMC feeder cells and various T feeder cells and cultured for 21 days (condition 4).
For re-stimulation at day 7 of culture, the natural killer cells being cultured were counted and diluted to a density of 2-5×105 cells/mL with 1 vol% autoplasma-containing CellGro medium (Cellgenix), and PBMC and T feeder cells were diluted 3- to 10-fold and suspended in 1 vol % autoplasma-containing CellGro medium (Cellgenix), and then inactivated by irradiation with 2000 and 15000-30000 cGy of gamma-rays, respectively, in a gamma-ray irradiator. 500IU of IL-2 and 10ng/mL of OKT-3 were added to the medium, and the prepared two types of cells were co-cultured. Next, the cells were suspension-cultured up to day 21 while the cells were counted at intervals of 2-3 days and diluted to a density of 5-10×105 cells/mL with CellGro medium (Cellgenix) containing 500IU of IL-2 and 1 vol % autoplasma. At day 21 of suspension culture, natural killer cells were harvested.
The proliferation rates of natural killer cells cultured under condition 2 shown in Table 1 were compared. As a result, it was shown that the fold increase in the number of total nucleated cells was higher in the order of PBMCs (334-fold), H9 (358-fold) and HuT78 (2282-fold). The fold increase in the number of natural killer cells was 1257-fold for PBMCs, 2677-fold for H9, and 29455-fold for HuT78 (FIG. 1b).
In addition, the proliferation rates of natural killer cells cultured under condition 4 of Table 1 while performing first stimulation with PBMC feeder cells and second stimulation with PBMC feeder cells and each type of T feeder cells were compared. As a result, it was shown that the fold increase in the number of total nucleated cells was 1402-fold for PBMCs, 720-fold for H9, and 1393-fold for HuT78 (FIG. 1d). Other T cells showed an increase in cell number of about 500-fold.
As described above, it could be seen that, in 14-day culture and 21-day culture, H9 and HuT78 can be used as feeder cells that are very excellent in terms of proliferation rate compared to PBMCs.
The results of evaluation performed using CD3(-) cells as seed cells and various T cells as feeder cells did slightly differ from the results obtained using PBMCs as feeder cells. Specifically, the proliferation rate of natural killer cells was similar between the PBMC feeder cells and the HuT78 feeder cells, and H9 feeder cells showed a reduced proliferation rate of natural killer cells compared to the PBMC feeder cells.
2-3: Culture of Poorly Proliferating Donor Cells
Donor cells that showed poor proliferation when cultured with PBMC feeder cells under condition 4 of Table 1 were selected and used to compare the proliferation rates of natural killer cells.
As a result, it was shown that culture with PBMC feeder cells showed an average of a 21-fold increase in the number of natural killer cells, but H9 and HuT78 showed a 64-fold increase and a 161-fold increase, respectively. Particularly, HuT78 showed an increase in cell number, which was about 8 times higher than that shown by PBMC feeder cells (FIG. 1e). Thus, it could be seen that T feeder cells can overcome the difference between donors with respect to the proliferation of natural killer cells, unlike PBMC feeder cells.
In the case of PBMC feeder cells, a process of depleting T cells from seed cells is necessarily required, because the proliferation of T cells rather than natural killer cells is induced when T cells are present in seed cells. In addition, a preculture process for previously selecting donors is required, because the difference in proliferation rate between donors is significant. However, it was found that T cells, particularly CD4-expressing T cells, could induce the selective proliferation of natural killer cells regardless of the presence or absence of T cells in seed cells and that the proliferation of poorly proliferating donor cells could also be easily induced when culture with PBMC feeder cells was performed.
Example 3: In Vitro Cell Viability
Among cell counter systems employing a PI dye capable of binding to the intracellular nucleus, an ADAM cell counter system was used to comparatively evaluate in vitro cell viabilities. The number of viable cells was determined by subtracting the measured dead cell count from the measured total cell count, and then cell viability was calculated using the following equation.
Cell viability (%) = (viable cell count/total cell count) × 100
The viability of natural killer cells was measured after culture under conditions 1 and 2 (that is, 14-day culture and 21-day culture of PBMC seed cells with various feeder cells) shown in Table 1. As a result, it was shown that the use of PBMC feeder cells showed a viability lower than 80%, and H9 and HuT78 showed a viability of 80% or higher (FIGS. 2a and 2b).
The viability of natural killer cells was measured after culture under conditions 3 and 4 (that is, 14-day culture and 21-day culture of CD3(+) T cell-depleted seed cells with various T feeder cells) shown in Table 1 were measured. As a result, it was shown that a high viability of about 90% or higher was shown in all the conditions. In the case of poorly proliferating donor cells, PBMC feeder cells showed a viability of 82%, and HuT78 showed a viability of 93% which was at least 10% higher than that shown by PBMC feeder cells (FIGS. 2c and 2d).
The viability of the cells cultured using PBMCs and various T feeder cells for 14 days or 21 days was generally higher in CD3(-) seed cells than in PBMC seed cells, and showed no significant difference between the types of seed cells. However, in the case of poorly proliferating donor cells, HuT78 showed a viability which was about 10% higher than that shown by PBMC feeder cells.
Thus, it could be seen that HuT78 cells are feeder cells for culturing natural killer cells, which are useful in terms of cell viability compared to PBMCs.
Example 4: In Vitro Analysis of Cell Phenotypes
Natural killer cells cultured according to the methods of Examples 1 and 2 were collected before and after culture and centrifuged at 1200 rpm for 5 minutes, and the medium was removed by suction. The cells were diluted with 1 mL of FACS buffer (2.5% FBS-containing PBS), counted, and then diluted to a density of 5×106 cells/mL with FACS buffer. 100L of the diluted cell solution was dispensed into each of 5mL FACS tubes (Falcon, 352052), and the phenotypes of the cells were analyzed using the following antibody:
Tube 1: anti-human CD3-FITC (BD Pharmingen, 555332), anti-human CD16-PE (BD Pharmingen, 555407), anti-human CD56-PE-Cy5 (BD Pharmingen, 555517)
Tube 2: anti-human CD14-FITC (BD Pharmingen, 555397), anti-human CD19-PE (BD Pharmingen, 555413), anti-human CD3-PE-Cy5 (BD Pharmingen, 555341)
Tube 3: anti-human CD3-FITC, anti-human NKG2A-PE (R&D system, FAB1059P), anti-human CD56-PE-Cy5
Tube 4: anti-human CD3-FITC, anti-human NKG2C-PE (R&D system, FAB138P), anti-humanCD56-PE-Cy5
Tube 5: anti-human CD3-FITC, anti-human NKG2D-PE (R&D system, FAB139P), anti-human CD56-PE-Cy5
Tube 6: anti-human CD3-FITC, anti-human NKp30-PE (BD Pharmingen, 558407), anti-human CD56-PE-Cy5
Tube 7: anti-human CD3-FITC, anti-human NKp44-PE (BD Pharmingen, 558563), anti-humanCD56-PE-Cy5
Tube 8: anti-human CD3-FITC, anti-human NKp46-PE (BD Pharmingen, 557991), anti-human CD56-PE-Cy5
Tube 9: anti-human CD3-FITC, anti-human DNAM-1-PE (BD Pharmingen, 559789), anti-humanCD56-PE-Cy5
Tube 10: anti-human CD3-FITC, anti-human CD25-PE (BD Pharmingen, 555432), anti-human CD56-PE-Cy5
Tube 11: anti-human CD3-FITC, anti-human CD62L-PE (eBioscience, 12-0629-42), anti-human CD56-PE-Cy5
Tube 12: anti-human CD3-FITC, anti-human CD69-PE (R&D systems, FAB23591P), anti-human CD56-PE-Cy5
Tube 13: anti-human CD3-FITC, anti-human CXCR3-PE (BD Pharmingen, 557185), anti-human CD56-PE-Cy5
Tube 14: anti-human CD3-FITC, anti-human CD57-PE (BD Pharmingen, 560844), anti-human CD56-PE-Cy5
Tube 15: anti-human CD3-FITC, PE mouse IgG1 k isotype control (BD Pharmingen, 555749), anti-human CD56-PE-Cy5
Tube 16: FITC mouse IgG1 k isotype control (BD Pharmingen, 555748), PE mouse IgG1 k isotype control (BD Pharmingen, 555749), PE-Cy5 mouse IgG1 k isotype control (BD Pharmingen)
The tubes were stained at a cold temperature for 30 minutes, and the stained cells were added to 2mL of FACS buffer and centrifuged at 1500 rpm for 5 minutes. The supernatant was removed, and the remaining cells were added again to 2 mL of FACS buffer and centrifuged at 1500 rpm for 5 minutes. Next, the supernatant was removed, and the remaining cells were suspended in 300 μL of FACS buffer, and the phenotypes thereof were analyzed using FACS LSRII Fortessa (Becton Dickinson), thereby determining the identity and purity of the cells. The content was expressed as the contents of CD3(-)CD56(+) cells and CD16(+)CD56(+) cells, and the purity was measured with CD3(+) for T cells, CD14(+) for monocytes, and CD19(+) for B cells.
4-1: Cell Identity and Purity
The identity and purity of natural killer cells cultured under conditions 1 and 2 shown in Table 1 were analyzed. As a result, the content of natural killer cells, evaluated after culturing PBMC seed cells with various feeder cells for 14 days, was 15.9% for PBMCs, 73.3% for H9, and 83.3% for HuT78, and the content of natural killer cells, evaluated after 21 days of culture, was 17.4% for PBMCs, 61.1% for H9, and 83.5% for HuT78 (see FIGS. 3a and 3b). Thus, it could be seen that, when PBMCs containing T cells were used as seed cells, in the case of PBMC feeder cells, T cells proliferated 80% or more, whereas in the case of H9 and HuT78, natural killer cells other than T cells selectively proliferated. Particularly, HuT78 showed a high natural killer cell content of 80% or more.
The identity and purity of natural killer cells cultured under conditions 3 and 4 shown in Table 1 were analyzed. As a result, it was shown that, because T cell-depleted CD3(-)seed cells were used under both the conditions, all the types of feeder cells showed a high natural killer cell content of 95% or more, and the contents of T cells, monocytes and B cells were all measured to be 1% or less (FIGS. 3c and 3d).
In addition, in the case of poorly proliferating donor cells cultured under condition 3 shown in Table 1, it was shown that culture with PBMC feeder cells showed a decrease in purity of about 4% and a decrease in CD16 expression of about 16%, compared to culture with HuT78 (FIG. 3e). Thus, it could be seen that CD4-expressing T cells could induce the selective proliferation of natural killer cells only and can significantly overcome the difference in culture of natural killer cells between donors.
4-2: Cell Expression Markers
In addition to the identity and purity of cells, the expression of typical natural killer cell receptors, which differs depending on the type of feeder cells, was analyzed.
The cell phenotypes of natural killer cells cultured under condition 1 of Table 1 were analyzed. As a result, for NKp44, NKp46 and CD69, the difference in phenotype between culture conditions was observed (FIG. 4a).
In addition, the cell phenotypes of natural killer cells cultured under condition 3 of Table 1 were analyzed. As a result, it was shown that the expression of NKp44 and NKp46 was higher in culture with H9 and HuT78 than in culture with PBMC and that the expression of CD69 was similar between culture with PBMC and culture with HuT78, but increased in culture with H9 (FIG. 4b).
Thus, it could be seen that, although the expression of cell phenotype markers did differ depending on the type of seed cells, the expression of natural killing cell activation markers such as NKp44 and NKp46 was significantly higher in culture with T feeder cells than in culture with PBMC feeder cells. These markers are important factors in the activity of natural killer cells, and the above results indicate that the efficacy of natural killer cells is higher in culture with T cells than in culture with PBMC feeder cells.
Example 5: In Vitro Cell Killing Activity Against Various Tumor Cell Lines
1×106 cells of each of target tumor cell lines (K562, HuT78, HuH-7, etc.) were dispensed into each of 15mL tubes and centrifuged. Then, the cell pellets were suspended in 1mL of RPMI 1640-10% FBS medium, after which 30μL of 1 mM Calcein-AM (Molecular probe, C34852) was added thereto, and then the cells were stained in an incubator at 37℃ for 1 hour while light was blocked with silver paper. The tumor cell line stained with Calcein-AM was washed with 10-15 mL of RPMI 1640-10% FBS medium and centrifuged, and the pellets were suspended in 10 mL of RPMI medium to a concentration of 1x105 cells/mL.
3×106 natural killer cells were dispensed into each of 15mL tubes and centrifuged, and the pellets were suspended in RPMI 1640-10% FBS medium at a desired ratio relative to the target tumor cell line. 100μL of each of the prepared target tumor cell line and natural killer cell line was dispensed into each well of a round-bottom 96-well plate, and each well was prepared in triplicate. To each spontaneous release well, 100μL of the stained tumor cell line and 100μL of RPMI 1640-10% FBS medium were added. To each maximum release well, 100μL of the stained tumor cell line and 100μL of 2% Triton-X 100 solution were added. To correct an autofluorescence value present in RPMI 1640-10% FBS medium and 2% Triton-X 100 solution, 200μL of RPMI 1640-10% FBS medium was added to prepare a medium value, 100μL of 2% Triton-X 100 solution was added to 100μL of RPMI 1640-10% FBS medium to prepare the value of the mixture of the two solutions. The difference (A) obtained by subtracting the value of the mixture from the medium value was added to the maximum release value, thereby correcting the autofluorescence value.
The cells were incubated in an incubator at 37℃ for 4 hours under a light-shielding condition, and then the plate was centrifuged at 2000 rpm for 3 minutes. 100 μL of the supernatant was added to each well of a 96-well black plate, and the fluorescence value (OD480/ 535 nm) was measured using a fluorescence plate reader (Perkin Elmer, VICTOR X3). Based on the measured fluorescence value, the tumor cell killing activity of the natural killer cells was calculated using the following equation:
% of killing = (Average fluorescence value of sample wells ― average fluorescence value of spontaneous wells)/{(average fluorescence value of maximum wells + A) ― average fluorescence value of spontaneous wells} × 100
Natural killer cells cultured with various feeder cells were allowed to react with various tumor cell lines, and the direct tumor cell killing activity of the natural killer cells was measured.
The cell killing activity of natural killer cells cultured under condition 1 (14-day culture of PBMC seed cells with various feeder cells) shown in Table 1 was evaluated against the blood cancer cell line K562, the liver cancer cell line HuH-7, the lymphoma cell line HuT78, the brain tumor cell line U87-MG, the retinoblastoma cell line SNUOT-Rbl, the neuroblastoma cell line SK-N-SH and the ovarian cancer cell line OVCAR-3. As a result, it was shown that culture with PBMC feeder cells showed a higher cell killing activity against all the tumor targets compared with culture with H9 and HuT78 (FIG. 5a).
The cell killing activity of natural killer cells cultured under condition 2 (21-day culture of PBMC seed cells) shown in Table 1 was measured. As a result, it was shown that the cell killing activity against the K562 tumor cell line was higher in the order of HuT78>H9>PBMCs (FIG. 5b).
In addition, the cell killing activity of natural killer cells cultured under condition 3 (14-day culture of CD3(-) seed cells with various feeder cells) was evaluated against the blood cancer cell line K562, the liver cancer cell line HuH-7, the lymphoma cell line HuT78, the brain tumor cell line U87-MG, the retinoblastoma cell line SNUOT-Rbl, the neuroblastoma cell line SK-N-SH and the ovarian cancer cell line OVCAR-3. As a result, it was shown that culture with H9 and HuT78 showed a higher cell killing activity against most of the tumor targets (excluding K562) compared to culture with PBMC feeder cells, and particularly, this difference was more significant in the case of cancer cells having resistance to the killing activity of natural killer cells (FIG. 5c).
The cell killing activity of natural killer cells cultured under condition 4 (21-day culture of CD3(-) seed cells) shown in Table 1 was measured. As a result, it was shown that the cell killing activity against the K562 tumor cell line was similar between the types of feeder cells (FIG. 5d). This insignificant difference in specificity between the types of feeder cells appears to be because PBMCs were used in the first stimulation of seed cells in the same manner and PBMCs and various T cells were used only in the second stimulation.
The cell killing activity of natural killer cells in the case of poorly proliferating donor cells, cultured under condition 3 of Table 3, was measured. As a result, it was shown that HuT78 showed the highest killing activity against the K562 tumor cell line and that H9 and PBMC feeder cells showed similar killing activities (FIG. 5e).
In conclusion, HuT78 showed the highest cell killing activity against various tumor cell lines under most of the conditions. Particularly, natural killer cells cultured with H9 and HuT78 showed higher killing activities against tumor cell lines having resistance, indicating that H9 and HuT78 cells are feeder cells which are better than PBMCs in terms of efficacy.
The characteristics of natural killer cells cultured under conditions 1, 2, 3 and 4 are summarized in the following Tables 2 to 5.
Figure PCTKR2015012700-appb-T000002
Figure PCTKR2015012700-appb-T000003
Figure PCTKR2015012700-appb-T000004
Figure PCTKR2015012700-appb-T000005
As described above, the method for producing natural killer cells using T cells according to the present invention is a method capable of producing natural killer cells by selectively proliferating only natural killer cells from a small amount of seed cells while maintaining the high killing activity of the natural killer cells. The method of the present invention can produce a large amount of natural killer cells that can be frozen, and thus is useful for commercialization of cell therapeutic agents.
Although the present invention has been described in detail with reference to the specific features, it will be apparent to those skilled in the art that this description is only for a preferred embodiment and does not limit the scope of the present invention. Thus, the substantial scope of the present invention will be defined by the appended claims and equivalents thereof.

Claims (23)

  1. A method of preparing natural killer cell comprising culturing seed cells by using CD4(+) T cells as feeder cells.
  2. The method of claim 1, wherein the CD4(+) T cells are CD4(+) T cells isolated ex vivo, CD4(+) T cells expansion-cultured ex vivo, or a CD4(+) T cell line.
  3. The method of claim 2, wherein the CD4(+) T cell line is H9 or HuT78.
  4. The method of claim 1, wherein the CD4(+) T cells are inactivated whose division and proliferation are inhibited, or non-inactivated cells.
  5. The method of claim 1, wherein the seed cells are one or more types selected from the group consisting of peripheral blood cells, peripheral blood leukocytes, PBMCs (peripheral blood mononuclear cells), enriched natural killer cells, and isolated natural killer cells.
  6. The method of claim 5, wherein the seed cells are CD3(+) cell-depleted cells.
  7. The method of claim 1, wherein the culturing is carried out by mixing feeder cells and the seed cells with a ratio of at least 1:1.
  8. The method of claim 7, wherein the culturing is carried out by mixing feeder cells and the seed cells with a ratio of 2:1-20:1.
  9. The method of claim 1, wherein the culturing is carried out for 5-60 days.
  10. The method of claim 1, wherein the culturing is carried out in a medium containing a T cell-stimulating antibody, which has a low affinity for T cells, and an interleukin protein.
  11. The method of claim 10, wherein the antibody is at least one selected from the group consisting of OKT3, UCHT1 and HIT3a antibodies.
  12. The method of claim 10, wherein the interleukin protein is at least one selected from the group consisting of IL-2, IL-12, IL-15, IL-18, and IL-21.
  13. The method of claim 1, further comprising re-stimulating CD3(+) cell-depleted seed cells by using 2~20 times CD4(+) T cells as feeder cells, compared to the seed cell.
  14. The method of claim 13, wherein the re-stimulation is performed at intervals of 5-12 days.
  15. The method of claim 13, wherein the re-stimulation is repeated at least once.
  16. The method of claim 13, wherein the CD4(+) T cells used in the re-stimulation step are H9 or HuT78.
  17. The method of claim 13, wherein the CD4(+) T cells used in the re-stimulation step are inactivated whose division and proliferation are inhibited, or non-inactivated cells.
  18. The method of claim 13, the method comprising culturing seed cells in a medium containing a T cell-stimulating antibody, which has a low affinity for T cells, and an interleukin protein.
  19. The method of claim 18, wherein the antibody is at least one selected from the group consisting of OKT3, UCHT1 and HIT3a antibodies.
  20. The method of claim 18, wherein the interleukin protein is at least one selected from the group consisting of IL-2, IL-12, IL-15, IL-18, and IL-21.
  21. Natural killer cells produced by any one of the methods of claims 1 to 20.
  22. A composition for preventing or treating cancer or infectious disease, which comprises the natural killer cells of claim 21 as an active ingredient.
  23. A method for preventing or treating cancer or infectious disease, comprising administering to a subject in need thereof a therapeutically effective amount of a composition comprising the natural killer cells of claim 21 as an active ingredient.
PCT/KR2015/012700 2014-11-26 2015-11-25 Method for culturing natural killer cells using t cells WO2016085248A1 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
US15/527,752 US20170319621A1 (en) 2014-11-26 2015-11-25 Method for culturing natural killer cells using t cells
EP15862930.3A EP3224349B1 (en) 2014-11-26 2015-11-25 Method for culturing natural killer cells using t cells
EP20216695.5A EP3878950A1 (en) 2014-11-26 2015-11-25 Method for culturing natural killer cells using t cells
JP2017525394A JP6720168B2 (en) 2014-11-26 2015-11-25 Method for culturing natural killer cells using T cells
CN201580063858.5A CN107002039B (en) 2014-11-26 2015-11-25 Method for culturing natural killer cells using T cells
ES15862930T ES2860974T3 (en) 2014-11-26 2015-11-25 Method for growing natural killer cells using T cells
AU2015354941A AU2015354941B2 (en) 2014-11-26 2015-11-25 Method for culturing natural killer cells using T cells
US17/220,865 US11766456B2 (en) 2014-11-26 2021-04-01 Method for culturing natural killer cells using T cells
US18/455,003 US20240050478A1 (en) 2014-11-26 2023-08-24 Method for Culturing Natural Killer Cells Using T Cells

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2014-0166705 2014-11-26
KR1020140166705A KR101697473B1 (en) 2014-11-26 2014-11-26 Method for Preparation of Natural Killer Cells Using T Cells

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US15/527,752 A-371-Of-International US20170319621A1 (en) 2014-11-26 2015-11-25 Method for culturing natural killer cells using t cells
US17/220,865 Continuation US11766456B2 (en) 2014-11-26 2021-04-01 Method for culturing natural killer cells using T cells

Publications (1)

Publication Number Publication Date
WO2016085248A1 true WO2016085248A1 (en) 2016-06-02

Family

ID=56074695

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2015/012700 WO2016085248A1 (en) 2014-11-26 2015-11-25 Method for culturing natural killer cells using t cells

Country Status (8)

Country Link
US (3) US20170319621A1 (en)
EP (2) EP3878950A1 (en)
JP (1) JP6720168B2 (en)
KR (1) KR101697473B1 (en)
CN (1) CN107002039B (en)
AU (1) AU2015354941B2 (en)
ES (1) ES2860974T3 (en)
WO (1) WO2016085248A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110799201A (en) * 2017-02-15 2020-02-14 加利福尼亚大学董事会 Compositions and methods for activating NK cells
JP2020521462A (en) * 2017-05-26 2020-07-27 グリーン・クロス・ラブ・セル・コーポレイション Method for culturing natural killer cells using transformed T cells
CN111902533A (en) * 2018-03-23 2020-11-06 Gc细胞治疗 Method for producing natural killer cells
US20200384082A1 (en) * 2017-12-14 2020-12-10 Green Cross Wellbeing Corporation Cosmetic composition and pharmaceutical composition for alleviating atopic dermatitis, hair loss, and wounds or reducing skin wrinkles
US11766456B2 (en) 2014-11-26 2023-09-26 GC Cell Corporation Method for culturing natural killer cells using T cells
US11970547B2 (en) 2017-11-14 2024-04-30 GC Cell Corporation Anti-HER2 antibody or antigen-binding fragment thereof, and chimeric antigen receptor comprising same

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102069704B1 (en) * 2018-05-16 2020-01-23 고려대학교 산학협력단 Method for Expansion of Human NK Cell Using HDAC Inhibitor
KR20200030337A (en) * 2018-09-12 2020-03-20 주식회사 녹십자랩셀 Pharmaceutical combinations for treating tumor comprising anti-cd19 antibody and natural killer cell
EP3892721A4 (en) * 2018-11-14 2022-08-31 GC Cell Corporation Method for culturing cord blood-derived natural killer cells using transformed t-cells
WO2020101361A1 (en) * 2018-11-14 2020-05-22 주식회사 녹십자랩셀 Method for culturing cord blood-derived natural killer cells using transformed t-cells
KR102216710B1 (en) * 2019-03-27 2021-02-17 신지섭 NK cell culture medium, NK cell culture method using the above-mentioned additive composition, and cosmetic composition for skin trouble improvement obtained by the above culture method
KR102495647B1 (en) * 2019-06-17 2023-02-06 라정찬 Method for Promoting Content of NK Cell Using Cultured Media of Immune Cell of Healthy Person
CN110195041B (en) * 2019-06-20 2023-04-25 威海正生生物科技有限公司 Method for obtaining high-activity Tregs cells by using ASC three-dimensional culture system
CN111518765B (en) * 2020-05-12 2021-01-26 优睿赛思(武汉)生物科技有限公司 B lymphocyte in-vitro culture system and application

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070048290A1 (en) 2005-08-31 2007-03-01 Schickwann Tsai Development of natural killer cells and functional natural killer cell lines
KR101035556B1 (en) * 2008-05-22 2011-05-23 재단법인 목암생명공학연구소 Method of preparing natural killer cells, natural killer cells prepared by the method, and composition for treatment of solid cancer containing same
US20120045423A1 (en) 2010-08-20 2012-02-23 Immunovative Therapies Ltd. Cells expressing TH1 characteristics and cytolytic properties
KR101133185B1 (en) 2008-07-29 2012-04-06 서울대학교병원 Method for Proliferating Natural Killer cell
KR20120091012A (en) * 2009-09-11 2012-08-17 다카라 바이오 가부시키가이샤 Process for production of natural killer cells
JP2013071915A (en) * 2011-09-28 2013-04-22 Biotherapy Institute Of Japan Method for producing nk cell strengthening type blood product
WO2013094988A1 (en) 2011-12-22 2013-06-27 Mogam Biotechnology Research Institute Method for producing natural killer cells, natural killer cells produced thereby, and composition for treating cancers and infectious diseases containing the same
KR101298012B1 (en) * 2011-02-08 2013-08-26 (주)차바이오앤디오스텍 Process for preparing lymphocytes comprising activated natural killer cells for targeting cancer cells and pharmaceutical composition comprising the same

Family Cites Families (46)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS5840837U (en) 1981-09-12 1983-03-17 三菱電機株式会社 Wafer cleaning and drying equipment
US5766601A (en) 1990-08-08 1998-06-16 University Of Massachusetts Medical Center Cross-reactive influenza a immunization
EP0852618A1 (en) 1995-07-25 1998-07-15 Celltherapy Inc. Autologous immune cell therapy: cell compositions, methods and applications to treatment of human disease
CA2247131A1 (en) 1996-03-04 1997-09-12 Targeted Genetics Corporation Modified rapid expansion methods ("modified-rem") for in vitro propagation of t lymphocytes
EP0947581A4 (en) 1996-08-08 2004-07-28 Mitsubishi Pharma Corp Culture medium and use of the same
MXPA02011587A (en) * 2000-05-23 2004-07-30 Nexell Therapeutics Inc Reagents for cell selection and methods of use.
US20030068306A1 (en) 2001-09-14 2003-04-10 Dilber Mehmet Sirac Medium
WO2005007116A2 (en) 2003-07-18 2005-01-27 Valeocyte Therapies Llc Artificial antigen presenting cell devices
PT1739166E (en) 2005-07-01 2011-09-22 Inst Nat Sante Rech Med Obtention of food- or auto-antigen specific tr1 cells from a leukocyte or pbmc population
US7628528B2 (en) 2005-10-26 2009-12-08 PRS Biotech, Inc. Pneumatic bioreactor
US20080261299A1 (en) 2007-04-23 2008-10-23 Zeikus J Gregory Pneumatic Bioreactor
KR20080008060A (en) 2006-07-19 2008-01-23 전남대학교산학협력단 A method for production of natural killer cell
JP2010501173A (en) 2006-08-23 2010-01-21 バイネックス カンパニー リミテッド Method for producing activated lymphocytes for immunotherapy
US20080138833A1 (en) 2006-12-07 2008-06-12 Wyeth Methods and compositions for assessing il-12 or the neutralization of il-12 in a sample
EP2141997B1 (en) 2007-03-30 2012-10-31 Memorial Sloan-Kettering Cancer Center Constitutive expression of costimulatory ligands on adoptively transferred t lymphocytes
CN101302491B (en) 2007-05-09 2011-09-14 王歈 Highly effective method for amplifying activated lymphocyte and cultivation system
WO2009060865A1 (en) 2007-11-05 2009-05-14 Tsuneo Kuramochi Pharmaceutical composition and method for production of pharmaceutical composition
US20090269849A1 (en) 2008-04-25 2009-10-29 Pbs Biotech, Inc. Bioreactor Apparatus
KR20090127974A (en) 2008-06-10 2009-12-15 주식회사 엔케이바이오 A medium composition for cultivating self activated lymphocyte
KR101043588B1 (en) 2008-07-25 2011-06-22 재단법인 철원플라즈마 산업기술연구원 Method Of Forming Ceramic Coating Layer having the Resistant Plasma
CN102428173B (en) 2009-03-26 2014-07-30 细胞维护北欧制药公司 Expansion of NK cells
EP3184109B1 (en) 2009-12-29 2020-11-18 Gamida-Cell Ltd. Methods for enhancing natural killer cell proliferation and activity
WO2012128622A1 (en) 2011-03-18 2012-09-27 Ipd-Therapeutics B.V. Generation of nk cells and nk-cell progenitors
EP2702145A1 (en) 2011-04-28 2014-03-05 Institut National de la Sante et de la Recherche Medicale (INSERM) Methods for preparing accessory cells and uses thereof for preparing activated nk cells
JP5840876B2 (en) 2011-06-24 2016-01-06 テラ株式会社 Compositions and methods for amplifying NK cells
JP5572863B2 (en) 2011-06-24 2014-08-20 国立大学法人九州大学 Method for amplifying NK cells
CN102559600A (en) 2011-12-29 2012-07-11 上海交通大学医学院 Artificial antigen presenting cell and application thereof in NK (natural killer) cell amplification
WO2013131045A1 (en) 2012-03-02 2013-09-06 The Regents Of The University Of California Expansion of alloantigen-reactive regulatory t cells
KR101520534B1 (en) 2012-05-07 2015-05-21 고려대학교 산학협력단 Method for enrichment and expansion of natural killer cells derived from peripheral blood mononuclear cells
JP5511039B1 (en) 2013-05-22 2014-06-04 国立大学法人九州大学 Method for preparing NK cells
RU2752275C2 (en) 2014-04-10 2021-07-26 Сиэтл Чилдрен'С Хоспитал (Дба Сиэтл Чилдрен'С Ресёрч Инститьют) Method and compositions for cellular immunotherapy
CA2958578A1 (en) 2014-08-21 2016-02-25 The General Hospital Corporation Tumor necrosis factor superfamily and tnf-like ligand muteins and methods of preparing and using the same
KR101697473B1 (en) 2014-11-26 2017-01-18 주식회사 녹십자랩셀 Method for Preparation of Natural Killer Cells Using T Cells
KR101706524B1 (en) 2014-12-03 2017-02-14 주식회사 녹십자랩셀 Efficient Method for Preparing of Stable Natural Killer Cells
GB201503500D0 (en) 2015-03-02 2015-04-15 Ucl Business Plc Cell
MY189692A (en) 2015-05-07 2022-02-26 Memorial Sloan Kettering Cancer Center Anti-ox40 antibodies and methods of use thereof
CN108934158B (en) 2016-02-01 2022-11-15 Gc细胞治疗 Culture medium composition for cell cryopreservation and application thereof
CN106222141B (en) 2016-10-17 2018-10-19 湖南丰晖生物科技有限公司 NK cell culture fluids and cell culture processes
CA3061898A1 (en) 2016-12-28 2019-10-29 Green Cross Lab Cell Corporation Chimeric antigen receptor and natural killer cells expressing same
GB201706451D0 (en) 2017-04-24 2017-06-07 Imp Innovations Ltd Cancer treatment
US20200108096A1 (en) 2017-05-26 2020-04-09 Green Cross Lab Cell Corporation Method for culturing natural killer cell, using transformed t cell
US20210179733A1 (en) 2017-11-14 2021-06-17 Green Cross Lab Cell Corporation Anti-her2 antibody or antigen-binding fragment thereof, and chimeric antigen receptor comprising same
WO2019182392A1 (en) 2018-03-23 2019-09-26 주식회사 녹십자랩셀 Method for producing natural killer cells
KR102232321B1 (en) 2018-03-23 2021-03-26 주식회사 녹십자랩셀 Method for Preparing NK Cell
WO2020101361A1 (en) 2018-11-14 2020-05-22 주식회사 녹십자랩셀 Method for culturing cord blood-derived natural killer cells using transformed t-cells
US20240000840A1 (en) 2020-12-17 2024-01-04 Artiva Biotherapeutics, Inc. Treatment of cancer with nk cells and a cd20 targeted antibody

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070048290A1 (en) 2005-08-31 2007-03-01 Schickwann Tsai Development of natural killer cells and functional natural killer cell lines
KR101035556B1 (en) * 2008-05-22 2011-05-23 재단법인 목암생명공학연구소 Method of preparing natural killer cells, natural killer cells prepared by the method, and composition for treatment of solid cancer containing same
KR101133185B1 (en) 2008-07-29 2012-04-06 서울대학교병원 Method for Proliferating Natural Killer cell
KR20120091012A (en) * 2009-09-11 2012-08-17 다카라 바이오 가부시키가이샤 Process for production of natural killer cells
US20120045423A1 (en) 2010-08-20 2012-02-23 Immunovative Therapies Ltd. Cells expressing TH1 characteristics and cytolytic properties
KR101298012B1 (en) * 2011-02-08 2013-08-26 (주)차바이오앤디오스텍 Process for preparing lymphocytes comprising activated natural killer cells for targeting cancer cells and pharmaceutical composition comprising the same
JP2013071915A (en) * 2011-09-28 2013-04-22 Biotherapy Institute Of Japan Method for producing nk cell strengthening type blood product
WO2013094988A1 (en) 2011-12-22 2013-06-27 Mogam Biotechnology Research Institute Method for producing natural killer cells, natural killer cells produced thereby, and composition for treating cancers and infectious diseases containing the same
KR20140123503A (en) * 2011-12-22 2014-10-22 재단법인 목암생명공학연구소 Method For Producing Natural Killer Cells, Natural Killer Cells Produced Thereby, And Composition For Treating Cancers And Infectious Diseases Containing The Same

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
BERG ET AL., CYTOTHERAPY, vol. 11, no. 3, 2009, pages 341 - 355
CONDIOTTI ET AL., EXPERIMENTAL HEMATOL., vol. 29, no. 1, 2001, pages 104 - 113
DENMAN ET AL., PLOSONE, vol. 7, no. 1, 2012, pages e53611
FUJISAKI ET AL., CANCER RES., vol. 69, no. 9, 2009, pages 4010 - 4017
GONG ET AL., TISSUE ANTIGENS, vol. 76, no. 6, 2010, pages 467 - 475
GOODIER ET AL., J. IMMUNOL., vol. 165, no. 1, 2000, pages 139 - 147
LIM ET AL., CANCER RES., vol. 73, no. 8, 2013, pages 2598 - 6607
NORTH ET AL., J. IMMUNOL., vol. 178, no. 1, 2007, pages 85 - 94

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11766456B2 (en) 2014-11-26 2023-09-26 GC Cell Corporation Method for culturing natural killer cells using T cells
CN110799201A (en) * 2017-02-15 2020-02-14 加利福尼亚大学董事会 Compositions and methods for activating NK cells
EP3582856A4 (en) * 2017-02-15 2021-03-10 The Regents of the University of California Compositions and methods for activating nk cells
JP2020521462A (en) * 2017-05-26 2020-07-27 グリーン・クロス・ラブ・セル・コーポレイション Method for culturing natural killer cells using transformed T cells
EP3633029A4 (en) * 2017-05-26 2021-06-09 Green Cross Lab Cell Corporation Method for culturing natural killer cell, using transformed t cell
JP7039623B2 (en) 2017-05-26 2022-03-22 グリーン・クロス・ラブ・セル・コーポレイション Method of culturing natural killer cells using transformed T cells
US11970547B2 (en) 2017-11-14 2024-04-30 GC Cell Corporation Anti-HER2 antibody or antigen-binding fragment thereof, and chimeric antigen receptor comprising same
US20200384082A1 (en) * 2017-12-14 2020-12-10 Green Cross Wellbeing Corporation Cosmetic composition and pharmaceutical composition for alleviating atopic dermatitis, hair loss, and wounds or reducing skin wrinkles
CN111902533A (en) * 2018-03-23 2020-11-06 Gc细胞治疗 Method for producing natural killer cells
EP3770251A4 (en) * 2018-03-23 2021-12-22 Green Cross Lab Cell Corporation Method for producing natural killer cells

Also Published As

Publication number Publication date
AU2015354941A1 (en) 2017-06-01
US20210268025A1 (en) 2021-09-02
CN107002039A (en) 2017-08-01
EP3878950A1 (en) 2021-09-15
AU2015354941B2 (en) 2018-11-15
KR20160063114A (en) 2016-06-03
KR101697473B1 (en) 2017-01-18
US20240050478A1 (en) 2024-02-15
EP3224349B1 (en) 2020-12-23
US11766456B2 (en) 2023-09-26
JP6720168B2 (en) 2020-07-08
EP3224349A1 (en) 2017-10-04
CN107002039B (en) 2021-04-23
EP3224349A4 (en) 2018-06-27
US20170319621A1 (en) 2017-11-09
ES2860974T3 (en) 2021-10-05
JP2018501779A (en) 2018-01-25

Similar Documents

Publication Publication Date Title
WO2016085248A1 (en) Method for culturing natural killer cells using t cells
Carosella et al. HLA-G–dependent suppressor cells: diverse by nature, function, and significance
WO2016209021A1 (en) Method for proliferating natural killer cells and composition for proliferating natural killer cells
KR101520534B1 (en) Method for enrichment and expansion of natural killer cells derived from peripheral blood mononuclear cells
WO2013094988A1 (en) Method for producing natural killer cells, natural killer cells produced thereby, and composition for treating cancers and infectious diseases containing the same
WO2017142187A1 (en) Medium addition kit for culturing nk cells and nk cell culturing method using kit
Khan et al. Expanded human blood-derived γδT cells display potent antigen-presentation functions
US20090130144A1 (en) Direct vaccination of the bone marrow
WO2019182392A1 (en) Method for producing natural killer cells
JPH04325087A (en) Production of cd4+helper t cell
Sosman et al. Specific recognition of human leukemic cells by allogeneic T cell lines
Takahashi et al. Dendritic cells generated from human blood in granulocyte macrophage-colony stimulating factor and interleukin-7
KR101867942B1 (en) Method for enrichment and expansion of virus antigen-specific T cells
WO2017003153A1 (en) Method for producing natural killer cells from cord blood monocytes or cells derived therefrom
WO2019231243A1 (en) Feeder cell expressing ox40l and method for culturing natural killer cells using same
Arstila et al. Helper activity of CD4+ αβ T cells is required for the avian γδ T cell response
WO2022060056A1 (en) Method for preparing high purity natural killer cells with high efficiency, and use thereof
Rabinowich et al. Clonal analysis of human tumor infiltrating lymphocytes reactive with autologous tumor cells: different target cell specificities of NK-like and cytotoxic T-cell clones
WO2020032782A1 (en) Method for preparing and cryopreserving cancer antigen-specific cd8+ t cells
KR101799986B1 (en) Method for Preparation of Natural Killer Cells Using T Cells
WO2023096352A1 (en) Feeder cell line genetically engineered to express hla-e and use thereof
Nunez Revision of the functional analysis and structural features of immortalized dendritic cell lines derived from mice lacking both type I and type II interferon receptors
Mou et al. 2B4 inhibits the apoptosis of natural killer cells through phosphorylated extracellular signal-related kinase/B-cell lymphoma 2 signal pathway
Yamamoto et al. Generation of lymphokine-activated killer cell activity by low-dose recombinant interleukin-2 and tumor cells
WO2023191559A1 (en) Method for mass proliferation of activated natural killer cells, and use thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 15862930

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2017525394

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 15527752

Country of ref document: US

REEP Request for entry into the european phase

Ref document number: 2015862930

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2015354941

Country of ref document: AU

Date of ref document: 20151125

Kind code of ref document: A